1
|
Ke Z, Lu Z, Li F, Zhao Q, Jiang X, Hu Z, Sun F, He Z, Tang Y, Li Q, van Oostendorp S, Chen X, He Q, Wang Y, Zhu Z, Tong W. Gut microbiota alterations induced by Roux-en-Y gastric bypass result in glucose-lowering by enhancing intestinal glucose excretion. Gut Microbes 2025; 17:2473519. [PMID: 40028693 PMCID: PMC11881838 DOI: 10.1080/19490976.2025.2473519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/26/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Roux-en-Y gastric bypass (RYGB) results in glucose-lowering in patients with type 2 diabetes mellitus (T2DM) and may be associated with increased intestinal glucose excretion. However, the contribution of intestinal glucose excretion to glycemic control after RYGB and its underlying mechanisms are not fully elucidated. Here, we confirmed that intestinal glucose excretion significantly increased in obese rats after RYGB, which was negatively correlated with postoperative blood glucose levels. Moreover, we also found that the contribution of Biliopancreatic limb length, an important factor affecting glycemic control after RYGB, to the improvement of glucose metabolism after RYGB attributed to the enhancement of intestinal glucose excretion. Subsequently, we further determined through multiple animal models that intestinal glucose excretion is physiological rather than pathological and plays a crucial role in maintaining glucose homeostasis in the body. Finally, we employed germ-free mice colonized with fecal samples from patients and rats to demonstrate that enhanced intestinal glucose excretion after RYGB is directly modulated by the surgery-induced changes in the gut microbiota. These results indicated that the gut microbiota plays a direct causal role in the hypoglycemic effect of RYGB by promoting intestinal glucose excretion, which may provide new insights for developing gut microbiota-based therapies for T2DM.
Collapse
Affiliation(s)
- Zhigang Ke
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Daping Hospital, Center for Hypertension and Metabolic Diseases, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Fan Li
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Qingyuan Zhao
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Xianhong Jiang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Zhihao Hu
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Daping Hospital, Center for Hypertension and Metabolic Diseases, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Zongcheng He
- Department of Digestive Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Tang
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Qing Li
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Stefan van Oostendorp
- Department of Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Qiuyue He
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Yong Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Daping Hospital, Center for Hypertension and Metabolic Diseases, Chongqing Institute of Hypertension, Army Medical University, Chongqing, China
| | - Weidong Tong
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
2
|
Cadena Sandoval M, Haeusler RA. Bile acid metabolism in type 2 diabetes mellitus. Nat Rev Endocrinol 2025; 21:203-213. [PMID: 39757322 DOI: 10.1038/s41574-024-01067-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/07/2025]
Abstract
Type 2 diabetes mellitus is a complex disorder associated with insulin resistance and hyperinsulinaemia that is insufficient to maintain normal glucose metabolism. Changes in insulin signalling and insulin levels are thought to directly explain many of the metabolic abnormalities that occur in diabetes mellitus, such as impaired glucose disposal. However, molecules that are directly affected by abnormal insulin signalling might subsequently go on to cause secondary metabolic effects that contribute to the pathology of type 2 diabetes mellitus. In the past several years, evidence has linked insulin resistance with the concentration, composition and distribution of bile acids. As bile acids are known to regulate glucose metabolism, lipid metabolism and energy balance, these findings suggest that bile acids are potential mediators of metabolic distress in type 2 diabetes mellitus. In this Review, we highlight advances in our understanding of the complex regulation of bile acids during insulin resistance, as well as how bile acids contribute to metabolic control.
Collapse
Affiliation(s)
- Marti Cadena Sandoval
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA
- Columbia Digestive and Liver Disease Research Center, Columbia University Medical Center, New York, NY, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA.
- Department of Medicine, Columbia University Medical Center, New York, NY, USA.
- Department of Pathology & Cell Biology, Columbia University Medical Center, New York, NY, USA.
- Columbia Digestive and Liver Disease Research Center, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
3
|
van der Kolk BW, Pirinen E, Nicoll R, Pietiläinen KH, Heinonen S. Subcutaneous adipose tissue and skeletal muscle mitochondria following weight loss. Trends Endocrinol Metab 2025; 36:339-363. [PMID: 39289110 DOI: 10.1016/j.tem.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/19/2024]
Abstract
Obesity is a major global health issue with various metabolic complications. Both bariatric surgery and dieting achieve weight loss and improve whole-body metabolism, but vary in their ability to maintain these improvements over time. Adipose tissue and skeletal muscle metabolism are crucial in weight regulation, and obesity is linked to mitochondrial dysfunction in both tissues. The impact of bariatric surgery versus dieting on adipose tissue and skeletal muscle mitochondrial metabolism remains to be elucidated. Understanding the molecular pathways that modulate tissue metabolism following weight loss holds potential for identifying novel therapeutic targets in obesity management. This narrative review summarizes current knowledge on mitochondrial metabolism following bariatric surgery and diet-induced weight loss in adipose tissue and skeletal muscle, and sheds light on their respective effects.
Collapse
Affiliation(s)
- Birgitta W van der Kolk
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Finland; Faculty of Medicine, Research Unit of Biomedicine and Internal Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Rachel Nicoll
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; HealthyWeightHub, Endocrinology, Abdominal Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Sini Heinonen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
4
|
Morissette A, Mulvihill EE. Cardioprotective benefits of metabolic surgery and GLP-1 receptor agonist-based therapies. Trends Endocrinol Metab 2025; 36:316-329. [PMID: 39127552 DOI: 10.1016/j.tem.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024]
Abstract
Individuals with excessive adipose tissue and type 2 diabetes mellitus (T2DM) face a heightened risk of cardiovascular morbidity and mortality. Metabolic surgery is an effective therapy for people with severe obesity to achieve significant weight loss. Additionally, metabolic surgery improves blood glucose levels and can lead to T2DM remission, reducing major adverse cardiovascular outcomes (MACE). Glucagon-like peptide 1 (GLP-1) receptor agonists (GLP-1RAs) are a class of medication that effectively reduce body weight and MACE in patients with T2DM. This review explores the potential mechanisms underlying the cardioprotective benefits of metabolic surgery and GLP-1RA-based therapies and discusses recent evidence and emerging therapies in this dynamic area of research.
Collapse
Affiliation(s)
- Arianne Morissette
- The University of Ottawa Heart Institute, 40 Ruskin Street, H-3229A, Ottawa, Ontario, KIY 4W7, Canada
| | - Erin E Mulvihill
- The University of Ottawa Heart Institute, 40 Ruskin Street, H-3229A, Ottawa, Ontario, KIY 4W7, Canada; Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, The University of Ottawa, 451 Smyth Road, Ottawa, Ontario, K1H 8L1, Canada.
| |
Collapse
|
5
|
Lowe AC, Reijnders D, Tam CS, Redman LM, Beyl R, LeBlanc KA, Hausmann MG, Albaugh VL, Greenway FL, Ravussin E. Changes in insulin sensitivity and gut peptides 8 and 52 weeks after bariatric surgery or low-calorie diet. Clin Obes 2025; 15:e12726. [PMID: 39688305 DOI: 10.1111/cob.12726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024]
Abstract
The endocrine consequences of weight loss by bariatric surgery (BS) and caloric restriction are not fully understood but contribute to variable improvements in insulin sensitivity and cardiometabolic health. This study compared changes in insulin sensitivity and plasma concentrations of gut peptides 8 weeks and 1 year after BS and a low-calorie diet (LCD). Nineteen female patients with obesity self-selected BS (gastric bypass [n = 5] or sleeve gastrectomy [n = 7]) or LCD (n = 7) in this parallel-arm, prospective observational study. We assessed insulin sensitivity via a two-step hyperinsulinemic-euglycemic clamp (20 and 80 mU/min/m2 insulin). Plasma glucose, insulin, and gut peptides were measured around a mixed meal tolerance test (400 kcal). Visual analogue scales (VAS) were used to rate subjective appetite sensations. All assessments were conducted at baseline and after 8 weeks and 1 year of intervention. Whole-body insulin sensitivity was unchanged 8 weeks after the intervention. One year after surgery, insulin sensitivity at both 20 and 80 mU/m2/min insulin infusion doses increased with BS weight loss (-33.8% ± 1.4% body weight) but was unchanged in LCD with small weight loss (-3.7% ± 2.0% body weight). Postprandial total PYY increased more following BS while total and acylated ghrelin decreased more following BS compared to LCD. Hunger decreased and fullness increased with BS compared to LCD (p = .037; p = .010, respectively). Insulin sensitivity was improved only 1 year after BS, despite significant weight loss after 8 weeks. Changes in gut peptides after BS paralleled reduced hunger and increased fullness. Most improvements in cardiometabolic health were related to weight loss.
Collapse
Affiliation(s)
- Adam C Lowe
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Dorien Reijnders
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Charmaine S Tam
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Leanne M Redman
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Robbie Beyl
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Karl A LeBlanc
- Our Lady of the Lake Physician Group, Franciscan Missionaries of Our Lady Health System, Baton Rouge, Louisiana, USA
| | - Mark G Hausmann
- Our Lady of the Lake Physician Group, Franciscan Missionaries of Our Lady Health System, Baton Rouge, Louisiana, USA
| | - Vance L Albaugh
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Frank L Greenway
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Eric Ravussin
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
6
|
Stentebjerg LL, Madsen LR, Støving RK, Hartmann B, Holst JJ, Vinter C, Juhl CB, Hojlund K, Jensen DM. Altered postprandial glucose metabolism and enteropancreatic hormone responses during pregnancy following Roux-en-Y gastric bypass: a prospective cohort study. BMJ Open Diabetes Res Care 2025; 13:e004672. [PMID: 40113260 PMCID: PMC11931895 DOI: 10.1136/bmjdrc-2024-004672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/08/2025] [Indexed: 03/22/2025] Open
Abstract
INTRODUCTION Roux-en-Y gastric bypass (RYGB) increases the risk of postprandial hypoglycemia, whereas pregnancy decreases insulin sensitivity, which could be expected to counteract hypoglycemia. We examined if RYGB performed prior to pregnancy altered the postprandial glucose metabolism and enteropancreatic hormone responses to a mixed meal test (MMT). RESEARCH DESIGN AND METHODS Twenty-three women with RYGB and 23 women matched on prepregnancy body mass index and parity underwent a 4-hour MMT in the first and third trimester of pregnancy with measurement of circulating levels of glucose, insulin, C-peptide, glucose-dependent insulin peptide (GIP), glucagon-like peptide 1 (GLP-1), glucagon, free fatty acids, and lactate. Biochemical hypoglycemia was defined as plasma glucose <3.5 mmol/L. RESULTS Women with RYGB had earlier and higher peak glucose, lower nadir glucose levels, and a higher frequency of biochemical hypoglycemia compared with women without RYGB in both the first and third trimester. The lower glucose levels were preceded by markedly elevated total GLP-1 and insulin levels in women with RYGB, whereas total GIP levels were unaltered. The glucagon levels were lower in women with RYGB. In the first trimester MMT, peak and area under the curve of total plasma GLP-1 and serum insulin levels were negatively associated with nadir plasma glucose, while the early postmeal response of plasma glucagon was positively associated with nadir plasma glucose in the third trimester. CONCLUSIONS These results provide novel insights into the combined effects of RYGB and pregnancy on postmeal glucose metabolism and enteropancreatic hormone responses during pregnancy, and how these changes associate with an increased risk of postprandial hypoglycemia. TRIAL REGISTRATION NUMBER NCT03713060.
Collapse
Affiliation(s)
- Louise Laage Stentebjerg
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lene Ring Madsen
- Steno Diabetes Center Aarhus, Aarhus Universitetshospital Skejby, Aarhus, Denmark
- Danish Diabetes Academy, Odense University Hospital, Odense, Denmark
| | - René Klinkby Støving
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Bolette Hartmann
- Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christina Vinter
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Gynecology and Obstetrics, Odense University Hospital, Odense, Denmark
| | - Claus Bogh Juhl
- Department of Endocrinology, University Hospital of Southern Denmark, Esbjerg, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Kurt Hojlund
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Dorte Møller Jensen
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Gynecology and Obstetrics, Odense University Hospital, Odense, Denmark
| |
Collapse
|
7
|
Orioli L, Thissen JP. Myokines as potential mediators of changes in glucose homeostasis and muscle mass after bariatric surgery. Front Endocrinol (Lausanne) 2025; 16:1554617. [PMID: 40171198 PMCID: PMC11958187 DOI: 10.3389/fendo.2025.1554617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Myokines are bioactive peptides released by skeletal muscle. Myokines exert auto-, para-, or endocrine effects, enabling them to regulate many aspects of metabolism in various tissues. However, the contribution of myokines to the dramatic changes in glucose homeostasis and muscle mass induced by bariatric surgery has not been established. Our review highlights that myokines such as brain-derived neurotrophic factor (BDNF), meteorin-like protein (Metrnl), secreted protein acidic and rich in cysteine (SPARC), apelin (APLN) and myostatin (MSTN) may mediate changes in glucose homeostasis and muscle mass after bariatric surgery. Our review also identifies myonectin as an interesting candidate for future studies, as this myokine may regulate lipid metabolism and muscle mass after bariatric surgery. These myokines may provide novel therapeutic targets and biomarkers for obesity, type 2 diabetes and sarcopenia.
Collapse
Affiliation(s)
- Laura Orioli
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
8
|
Schauer PR, Rothberg AE. Point-Counterpoint Debate: Surgery vs Medical Treatment for the Management of Obesity. J Clin Endocrinol Metab 2025; 110:e1282-e1287. [PMID: 39693237 DOI: 10.1210/clinem/dgae888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/20/2024]
Abstract
Obesity is a chronic, relapsing condition with severe health risks and a huge economic burden. Effective interventions for severe obesity include bariatric or metabolic surgery and high-intensity medical management involving lifestyle changes and pharmacotherapy. This article summarizes the debate between Drs. Schauer and Rothberg at the June 2024 Endocrine Society meeting regarding the optimal approach to managing obesity, exploring mechanisms, outcomes, safety, quality-of-life, and cost-effectiveness. Metabolic surgery results in substantial and sustained weight loss, improvements in comorbidities such as type 2 diabetes, and reduced mortality, and it is cost-effective. However, it carries risks associated with surgery and long-term complications, and its high upfront costs limit its scalability. Conversely, high-intensity medical management, which includes comprehensive lifestyle interventions and pharmacotherapy, leads to meaningful, though sometimes less substantial, weight loss and health improvements. The latter approach prioritizes behavioral changes and is cost-effective but requires patient adherence and faces challenges with medication side effects and costs. Both interventions offer substantial health benefits; the choice between them should consider individual patient needs, health status, target weight loss, and personal preferences. Metabolic surgery may be more suitable for individuals with severe obesity or when comorbidities are inadequately controlled, whereas medical management may be more suited to patients with less severe obesity and those preferring nonsurgical options. Future research should investigate the combined effects of surgery and medical management and enhance access to and affordability of these treatments. A multidisciplinary, personalized approach will likely yield the best outcomes in managing this complex health issue.
Collapse
Affiliation(s)
- Philip R Schauer
- Pennington Biomedical Research Institute of Louisiana State University in Baton Rouge, Baton Rouge, LA 70808, USA
| | - Amy E Rothberg
- Department of Internal Medicine, Michigan Medicine and Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
9
|
Podobnik J, Prentice KJ. Metabolic interventions as adjunctive therapies to insulin in type 1 diabetes: Current clinical landscape and perspectives. Diabetes Obes Metab 2025; 27:1032-1044. [PMID: 39757938 PMCID: PMC11802405 DOI: 10.1111/dom.16154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/04/2024] [Accepted: 12/14/2024] [Indexed: 01/07/2025]
Abstract
Type 1 diabetes (T1D) is classically characterized as an autoimmune disease wherein the immune system erroneously attacks insulin-producing pancreatic β-cells, causing insulin insufficiency and severe metabolic dysregulation. However, intensive investigation and numerous clinical trials with immunotherapies have been largely unable to significantly alter the course of disease. Currently, there is no effective way to prevent or cure T1D, and insulin remains the cornerstone of T1D treatment. In recent years, a growing body of research suggests that β-cells actively contribute to the immune response and to disease development. Factors including glucotoxicity, lipotoxicity, inflammation, endoplasmic reticulum (ER) and oxidative stress can induce β-cell apoptosis and senescence, further promoting insulitis. Recent studies highlight the importance of targeting metabolic control for T1D management and treatment. Metabolic interventions, through their direct and indirect impacts on β-cells, have shown promise in preserving β-cell function. These interventions can reduce glucose toxicity, alleviate oxidative stress and inflammation, enhance insulin sensitivity, and indirectly mitigate the autoimmune responses. By preserving β-cell function, individuals with T1D attain better glycaemic control, reduced complication risks and exhibit improved overall metabolic health. Here, we provide an overview of insights from clinical studies, systematic reviews and meta-analyses that collectively demonstrate that adjunctive metabolic interventions can enhance glycaemic control, reduce insulin requirements and mitigate adverse effects associated with insulin monotherapy. They also show potential for halting disease progression, preserving residual β-cell function and improving long-term outcomes for newly diagnosed individuals. Future research should focus on optimizing these treatment strategies and establishing their long-term efficacy and safety.
Collapse
Affiliation(s)
- Juliana Podobnik
- Department of Physiology, Temerty Faculty of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Kacey J. Prentice
- Department of Physiology, Temerty Faculty of MedicineUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
10
|
Taylor R. The Twin Cycle Hypothesis of type 2 diabetes aetiology: From concept to national NHS programme. Exp Physiol 2025. [PMID: 39898429 DOI: 10.1113/ep092009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
The development of magnetic resonance methods for quantifying intra-organ metabolites has permitted advances in the understanding of fasting and post-prandial carbohydrate and lipid handling in people with and without type 2 diabetes. Insulin resistance in the liver was shown to be related to excess intra-organ fat and was able to be returned to normal by weight loss. The practical effect of having muscle insulin sensitivity in the lower part of the wide normal range resulted in the obligatory shunting of carbohydrates via de novo lipogenesis into saturated fat. These observations provided the basis for the Twin Cycle Hypothesis of the aetiology of type 2 diabetes. Subsequent studies on people with type 2 diabetes confirmed the postulated pathophysiological abnormalities and demonstrated their reversibility by dietary weight loss of 10-15 kg. Overall, the fundamental understanding of the mechanisms causing type 2 diabetes has bridged physiological and clinical perspectives. Large population-based randomised controlled trials confirmed the practical clinical application of the method of achieving substantial weight loss, and an NHS programme is now in place offering potential remission to people within 6 years of diagnosis.
Collapse
Affiliation(s)
- Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
11
|
Simancas-Racines D, Annunziata G, Verde L, Fascì-Spurio F, Reytor-González C, Muscogiuri G, Frias-Toral E, Barrea L. Nutritional Strategies for Battling Obesity-Linked Liver Disease: the Role of Medical Nutritional Therapy in Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Management. Curr Obes Rep 2025; 14:7. [PMID: 39797961 PMCID: PMC11724794 DOI: 10.1007/s13679-024-00597-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/03/2024] [Indexed: 01/13/2025]
Abstract
PURPOSE OF REVIEW This narrative review explores the role of Medical Nutritional Therapy (MNT) in managing Metabolic-Associated Steatotic Liver Disease (MASLD), previously known as nonalcoholic fatty liver disease. It aims to examine the effectiveness of specific nutritional strategies in preventing and treating this obesity-linked liver disease. RECENT FINDINGS Emerging evidence underscores the benefits of the Mediterranean diet, low-carbohydrate diets, and intermittent fasting in reducing liver fat, improving insulin sensitivity, and mitigating inflammation. Supplementing with vitamin E, omega-3 fatty acids, and silymarin can potentially reduce liver fibrosis and promote liver health. MNT is a key intervention for MASLD management, emphasizing dietary patterns, caloric restriction, and nutraceutical supplementation. Integrating these strategies with lifestyle modifications, including regular physical activity, offers a comprehensive approach to improving metabolic and liver outcomes in patients with MASLD. Further research is needed to refine and personalize these therapeutic interventions.
Collapse
Affiliation(s)
- Daniel Simancas-Racines
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, 170527, Ecuador.
| | - Giuseppe Annunziata
- Facoltà di Scienze Umane, della Formazione e dello Sport, Università Telematica Pegaso, Via Porzio, Centro Direzionale, Isola F2, Naples, 80143, Italy
| | - Ludovica Verde
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Claudia Reytor-González
- Universidad UTE, Facultad de Ciencias de la Salud Eugenio Espejo, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Quito, 170527, Ecuador
| | - Giovanna Muscogiuri
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, 80131, Naples, Italy
| | - Evelyn Frias-Toral
- Universidad Espíritu Santo, Escuela de Medicina, Samborondón, 0901952, Ecuador.
| | - Luigi Barrea
- Dipartimento Psicologia e Scienze della Salute, Università Telematica Pegaso, Centro Direzionale Isola F2, Via Porzio, Naples, 80143, Italy
| |
Collapse
|
12
|
Song K, Kong X, Xian Y, Yu Z, He M, Xiao D, Liang D, Zhang Z, Liu T, Huang Z, Liao X, Ren Y. Roux-en-Y gastric bypass improves liver and glucose homeostasis in Zucker diabetic fatty rats by upregulating hepatic trefoil factor family 3 and activating the phosphatidylinositol 3-kinase/protein kinase B pathway. Surg Obes Relat Dis 2025:S1550-7289(25)00007-3. [PMID: 39893149 DOI: 10.1016/j.soard.2024.12.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/25/2024] [Accepted: 12/22/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) surgery is effective in ameliorating type 2 diabetes mellitus (T2DM); but its mechanism remains incompletely understood. OBJECTIVES This study aimed to investigate whether RYGB improves glucose metabolism by upregulating hepatic trefoil factor family 3 (TFF3) and thereby activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway. SETTING Affiliated Hospital of North Sichuan Medical college in Nanchong city, China. METHODS Zucker diabetic fatty (ZDF) rats underwent RYGB or sham surgery (SHAM), and Zucker lean (ZL) rats served as controls (CON). TFF3 expression and PI3K/Akt pathway activity were compared between groups using western blot, immunofluorescence, and RT-qPCR. Adeno-associated virus (AAV) was used to specifically overexpress and interfere with hepatic TFF3. Liver fibrosis and steatosis were assessed using Masson trichrome and Oil Red O staining. HepG2 cells overexpressing or knocking out TFF3 were constructed using lentiviral transfection and CRISPR/Cas9 technology. After verifying the activity of the PI3K/Akt pathway by western blot, rescue experiments were performed on HepG2 cell overexpressing and knocking out TFF3 using LY294002 and 740Y-P, respectively. The activities of gluconeogenic enzymes and glucose uptake capacity in different HepG2 cells were evaluated using qPCR and flow cytometry. RESULTS Compared with the SHAM group, the blood glucose, body weight, insulin resistance, and lipid metabolism of ZDF rats in the RYGB group were significantly improved. The expression of TFF3 and PI3K/Akt phosphorylation in the liver of the RYGB group were higher than those of the rats that had undergone SHAM. In addition, compared with the SHAM group, the liver fibrosis and fatty degeneration of RYGB rats were milder, and the activity of gluconeogenic enzymes was lower. After tail vein injection of AAV that specifically overexpresses liver TTF3 in rats in the SHAM group, rats' insulin resistance, glucose tolerance, gluconeogenic enzymes, and other glucose metabolism indicators improved. After tail vein injection of AAV that interferes with liver TFF3 in rats in the RYGB group, rats' glucose metabolism indicators deteriorated. In in vitro experiments, the PI3K/Akt activity of TFF3-knocked-out HepG2 cells was lower than that of other groups. Lower glucose concentration were observed in TFF3-overexpressing cell lines. After rescue experiments, differences were found. The glucose metabolism level of the TFF3-expressing HepG2 cell line was positively correlated with the activity of the PI3K/Akt pathway. CONCLUSIONS RYGB regulates the expression of TFF3 in the liver of ZDF rats, thereby activating the PI3K/Akt pathway and improving T2DM.
Collapse
Affiliation(s)
- Ke Song
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Xiangxin Kong
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Yin Xian
- Department of Otolaryngology & Head and Neck Surgery, Nanchong Psychosomatic Hospital, Nanchong, China
| | - Zhenghang Yu
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Ming He
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Dingqi Xiao
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Dianyuan Liang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Zhongyang Zhang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Ting Liu
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Ziyan Huang
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Xinxin Liao
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, China
| | - Yixing Ren
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China; General Surgery, Chengdu XinHua Hospital Affiliated to North Sichuan Medical College, Chengdu, China.
| |
Collapse
|
13
|
Sarıakçalı B, Şahin FN, Başoğlu B, Ceylan L, Güler Ö, Yamak B, Arıkan G, Acar GC, Sekban G, Durmuşoğlu MV, Çimen Polat S, Küçük H. The dual impact: physiological and psychological effects of rapid weight loss in wrestling. Front Psychol 2025; 15:1513129. [PMID: 39834765 PMCID: PMC11743537 DOI: 10.3389/fpsyg.2024.1513129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Athletes competing in weight-class sports often seek to gain an advantage by competing at lower weights. Athletes competing in weight-class sports often seek to gain an advantage by competing at lower weights. To achieve this, they aim to lose weight during the competition period, leading to various physiological and psychological changes. This study aimed to investigate the biochemical, hormonal, and psychological effects of weight reduction in elite wrestlers during the competition phase. Methods Thirty-seven elite male free style wrestlers (age: 19.02 ± 1.27) participated in the study. Samples were collected 5 days before and on the day of the match. Results A significant decrease in body weight was observed (p < 0.05). Levels of creatine, BUN, sodium, hematocrit, hemoglobin, LDH, and cortisol increased, while albumin, testosterone, and FSH levels decreased. There were no significant differences in potassium, ALT, AST, TSH levels. State and trait anxiety scores of the wrestlers increased significantly during the RWL period. Conclusion The study concluded that elite wrestlers experienced significant changes in physiological and psychological parameters during the competition periods. These findings underscore the importance of careful monitoring of RWL strategies by coaches and athletes to mitigate the adverse effects on nutritional status, psychological well-being, and physical performance.
Collapse
Affiliation(s)
- Barış Sarıakçalı
- Department of Internal Medicine, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Fatma Neşe Şahin
- Department of Coaching Education, Faculty of Sport Sciences, Ankara University, Ankara, Türkiye
| | - Burhan Başoğlu
- Department of Physical Education and Sport, Faculty of Sport Sciences, Nevşehir Haci Bektaş Veli University, Nevşehir, Türkiye
| | - Levent Ceylan
- Department of Sport Management, Faculty of Sport Sciences, Hitit University, Corum, Türkiye
| | - Özkan Güler
- Department of Coaching Education, Faculty of Sport Sciences, Ankara University, Ankara, Türkiye
| | - Bade Yamak
- Department of Recreation, Yasar Dogu Faculty of Sport Sciences, Ondokuz Mayis University, Samsun, Türkiye
| | - Gökhan Arıkan
- Mehmet Arabaci School of Physical Education and Sport, Harran University, Sanliurfa, Türkiye
| | - Gizem Ceylan Acar
- Department of Sport Management, Faculty of Sport Sciences, Mus Alsarslan University, Mus, Türkiye
| | - Gülşah Sekban
- Department of Physical Education and Sport, Faculty of Sport Sciences, Sinop University, Sinop, Türkiye
| | | | - Sezen Çimen Polat
- Department of Coaching Education, Faculty of Sport Sciences, Gazi University, Ankara, Türkiye
| | - Hamza Küçük
- Department of Physical Education and Sport, Yasar Dogu Faculty of Sport Sciences, Ondokuz Mayis University, Samsun, Türkiye
| |
Collapse
|
14
|
Petersen MC, Yoshino M, Smith GI, Gaspar RC, Kahn M, Samovski D, Shulman GI, Klein S. Effect of Weight Loss on Skeletal Muscle Bioactive Lipids in People With Obesity and Type 2 Diabetes. Diabetes 2024; 73:2055-2064. [PMID: 39264820 PMCID: PMC11579410 DOI: 10.2337/db24-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024]
Abstract
Muscle sn-1,2-diacylglycerol (DAG) and C18:0 ceramide accumulation in sarcolemmal and mitochondrial compartments have been proposed to regulate muscle insulin sensitivity. Here, we evaluated whether weight loss-induced improvements in insulin sensitivity were associated with changes in muscle sn-1,2-DAG and ceramide content in people with obesity and type 2 diabetes. We measured skeletal muscle insulin sensitivity, assessed by using the hyperinsulinemic-euglycemic clamp procedure in conjunction with stable isotopically labeled glucose tracer infusion, and skeletal muscle sn-1,2-DAG and ceramide contents by using liquid chromatography-tandem mass spectrometry after subcellular fractionation and DAG isomer separation in 14 adults with obesity and type 2 diabetes before and after marked (18.6 ± 2.1%) weight loss. Whole-body insulin sensitivity doubled after weight loss. Sarcolemmal sn-1,2-DAG and C18:0 ceramide contents after weight loss were not different from values before weight loss. In contrast, mitochondrial-endoplasmic reticulum (ER) C18:0 ceramide content decreased by ∼20% after weight loss (from 2.16 ± 0.08 to 1.71 ± 0.13 nmol/g, P < 0.005). These results suggest a decrease in muscle mitochondrial-ER C18:0 ceramide content could contribute to the beneficial effect of weight loss on skeletal muscle insulin sensitivity. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Max C. Petersen
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO
- Division of Endocrinology, Metabolism, & Lipid Research, Washington University in St. Louis, St. Louis, MO
| | - Mihoko Yoshino
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO
| | - Gordon I. Smith
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO
| | - Rafael C. Gaspar
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
| | - Mario Kahn
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
| | - Dmitri Samovski
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO
| | - Gerald I. Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT
| | - Samuel Klein
- Center for Human Nutrition, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
15
|
Taylor R. Similar early metabolic changes induced by dietary weight loss or bariatric surgery. Diabetologia 2024; 67:2603-2604. [PMID: 39235458 DOI: 10.1007/s00125-024-06268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 09/06/2024]
Affiliation(s)
- Roy Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
16
|
Mucinski JM, Salvador AF, Moore MP, Fordham TM, Anderson JM, Shryack G, Cunningham RP, Lastra G, Gaballah AH, Diaz-Arias A, Ibdah JA, Rector RS, Parks EJ. Histological improvements following energy restriction and exercise: The role of insulin resistance in resolution of MASH. J Hepatol 2024; 81:781-793. [PMID: 38914313 DOI: 10.1016/j.jhep.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatohepatitis (MASH) is one of the most common liver diseases worldwide and is characterized by multi-tissue insulin resistance. The effects of a 10-month energy restriction and exercise intervention on liver histology, anthropometrics, plasma biochemistries, and insulin sensitivity were compared to standard of care (control) to understand mechanisms that support liver health improvements. METHODS Following medical diagnosis of MASH, individuals were randomized to treatment (n = 16) or control (n = 8). Liver fat (magnetic resonance spectroscopy), 18-hour plasma biochemical measurements, and isotopically labeled hyperinsulinemic-euglycemic clamps were completed pre- and post-intervention. Body composition and cardiorespiratory fitness (VO2peak) were also measured mid-intervention. Those in the treatment group were counseled to reduce energy intake and completed supervised, high-intensity interval training (3x/week) for 10 months. Controls continued physician-directed care. RESULTS Treatment induced significant (p <0.05) reductions in body weight, fat mass, and liver injury, while VO2peak (p <0.05) and non-esterified fatty acid suppression (p = 0.06) were improved. Both groups exhibited reductions in total energy intake, hemoglobin A1c, hepatic insulin resistance, and liver fat (p <0.05). Compared to control, treatment induced a two-fold increase in peripheral insulin sensitivity which was significantly related to higher VO2peak and resolution of liver disease. CONCLUSIONS Exercise and energy restriction elicited significant and clinically meaningful treatment effects on liver health, potentially driven by a redistribution of excess nutrients to skeletal muscle, thereby reducing hepatic nutrient toxicity. Clinical guidelines should emphasize the addition of aerobic exercise in lifestyle treatments for the greatest histologic benefit in individuals with advanced MASH. IMPACT AND IMPLICATIONS The mechanisms that underpin histologic improvement in individuals with metabolic dysfunction-associated steatohepatitis (MASH) are not well understood. This study evaluated the relationship between liver and metabolic health, testing how changes in one may affect the other. We investigated the effects of energy restriction and exercise on the association between multi-tissue insulin sensitivity and histologic improvements in participants with biopsy-proven MASH. For the first time, these results show that an improvement in peripheral (but not hepatic) insulin sensitivity and systemic markers of muscle function (i.e. cardiorespiratory fitness) were strongly related to resolution of liver disease. Extrahepatic disposal of substrates and improved fitness levels supported histologic improvement, confirming the addition of exercise as crucial to lifestyle interventions in MASH. CLINICAL TRIAL NUMBER NCT03151798.
Collapse
Affiliation(s)
- Justine M Mucinski
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States
| | - Amadeo F Salvador
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States
| | - Mary P Moore
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States; Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO 65201, United States
| | - Talyia M Fordham
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States
| | - Jennifer M Anderson
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States
| | - Grace Shryack
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States; NextGen Precision Health, Columbia, MO 65201, United States
| | - Rory P Cunningham
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States; Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO 65201, United States
| | - Guido Lastra
- Endocrinology and Metabolism, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Ayman H Gaballah
- Department of Radiology, School of Medicine, University of Missouri, Columbia, MO, 65212, United States
| | - Alberto Diaz-Arias
- Boyce & Bynum Pathology Laboratories, Columbia, MO, 65201, United States
| | - Jamal A Ibdah
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States; Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO 65201, United States; Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Missouri, Columbia, MO 65212, United States; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, 65212, United States
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States; Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, MO 65201, United States; NextGen Precision Health, Columbia, MO 65201, United States; Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO 65212, United States; NextGen Precision Health, Columbia, MO 65201, United States; Department of Medicine, Division of Gastroenterology and Hepatology, School of Medicine, University of Missouri, Columbia, MO 65212, United States.
| |
Collapse
|
17
|
Axelrod CL, Hari A, Dantas WS, Kashyap SR, Schauer PR, Kirwan JP. Metabolomic Fingerprints of Medical Therapy Versus Bariatric Surgery in Patients With Obesity and Type 2 Diabetes: The STAMPEDE Trial. Diabetes Care 2024; 47:2024-2032. [PMID: 39311919 PMCID: PMC11502526 DOI: 10.2337/dc24-0859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/28/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVE Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) are effective procedures to treat and manage type 2 diabetes (T2D). However, the underlying metabolic adaptations that mediate improvements in glucose homeostasis remain largely elusive. The purpose of this study was to identify metabolic signatures associated with biochemical resolution of T2D after medical therapy (MT) or bariatric surgery. RESEARCH DESIGN AND METHODS Plasma samples from 90 patients (age 49.9 ± 7.6 years; 57.7% female) randomly assigned to MT (n = 30), RYGB (n = 30), or SG (n = 30) were retrospectively subjected to untargeted metabolomic analysis using ultra performance liquid chromatography with tandem mass spectrometry at baseline and 24 months of treatment. Phenotypic importance was determined by supervised machine learning. Associations between change in glucose homeostasis and circulating metabolites were assessed using a linear mixed effects model. RESULTS The circulating metabolome was dramatically remodeled after SG and RYGB, with largely overlapping signatures after MT. Compared with MT, SG and RYGB profoundly enhanced the concentration of metabolites associated with lipid and amino acid signaling, while limiting xenobiotic metabolites, a function of decreased medication use. Random forest analysis revealed 2-hydroxydecanoate as having selective importance to RYGB and as the most distinguishing feature between MT, SG, and RYGB. To this end, change in 2-hydroxydecanoate correlated with reductions in fasting glucose after RYGB but not SG or MT. CONCLUSIONS We identified a novel metabolomic fingerprint characterizing the longer-term adaptations to MT, RYGB, and SG. Notably, the metabolomic profiles of RYGB and SG procedures were distinct, indicating equivalent weight loss may be achieved by divergent effects on metabolism.
Collapse
Affiliation(s)
- Christopher L. Axelrod
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Adithya Hari
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Wagner S. Dantas
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | | | - Philip R. Schauer
- Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH
- Clinical Metabolic Surgery Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - John P. Kirwan
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
18
|
Aoyama K, Nakajima Y, Meguro S, Hayashi K. Effects of weight loss from oral semaglutide administration on cardiometabolic risk factors in Japanese patients with type 2 diabetes: a retrospective analysis using propensity score matching. Diabetol Int 2024; 15:794-805. [PMID: 39469553 PMCID: PMC11512971 DOI: 10.1007/s13340-024-00744-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/30/2024] [Indexed: 10/30/2024]
Abstract
Background Obesity is increasingly being recognized as a chronic disease that exacerbates type 2 diabetes and its related complications. Oral semaglutide, a novel glucagon-like peptide-1 (GLP-1) receptor agonist, has demonstrated efficacy in weight loss and diabetes control in Western populations. However, in real-world clinical practice, its effectiveness in Japanese patients, who typically exhibit a leaner phenotype and unique genetic susceptibilities affecting insulin secretion, remains unclear. Methods We retrospectively evaluated the electronic medical records of 313 patients treated with oral semaglutide and 11,239 untreated controls at the Keio University School of Medicine. We performed propensity score matching to adjust for covariates, including age, sex, height, weight, blood pressure, blood test data, medications, and compared the cardiometabolic risk factors, including HbA1c, blood pressure, lipids, and liver function 180 days post-treatment, of both patient groups. We conducted a subgroup analysis for patients who achieved ≥ 3% weight loss. Results After propensity score matching, the semaglutide group demonstrated significantly better outcomes for HbA1c reduction and weight loss and improvements in systolic blood pressure (SBP), low-density lipoprotein cholesterol (LDL-C), and liver function than the control group. Subgroup analysis of patients with ≥ 3% weight loss revealed superior HbA1c improvements in the semaglutide group; however, no significant differences in other metabolic parameters, such as SBP, LDL-C, and liver function, were observed. Conclusion Oral semaglutide effectively improved metabolic markers in Japanese patients with type 2 diabetes, similar to that in Western populations. Weight loss itself was suggested to significantly contribute to blood pressure, lipid levels, and liver function changes. Supplementary Information The online version contains supplementary material available at 10.1007/s13340-024-00744-3.
Collapse
Affiliation(s)
- Kazuki Aoyama
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Yuya Nakajima
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shu Meguro
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Kaori Hayashi
- Department of Endocrinology, Metabolism and Nephrology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
19
|
Ghusn W, Salameh Y, Abi Mosleh K, Shah M, Storm AC, Abu Dayyeh BK, Ghanem OM. Using Pre-operative Insulin Dose to Predict Diabetes Remission After Roux-En-Y Gastric Bypass and Sleeve Gastrectomy. Surg Innov 2024; 31:484-492. [PMID: 39105388 DOI: 10.1177/15533506241273368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
BACKGROUND Obesity is intricately associated with type-2 diabetes (T2D) and other cardiovascular conditions, increasing morbidity, mortality, and health care costs. Metabolic and bariatric surgeries (MBS) have shown promising results in significant weight loss and T2D remission, but existing predictive scores for post-MBS diabetes remission do not consider insulin dosage, potentially overlooking a critical factor. METHODS A retrospective analysis of patients with T2D who underwent Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG). The study focused on insulin dosage impact, divided into quartiles, on remission rates post-MBS. The effectiveness of RYGB vs SG was compared within insulin dose quartiles with up to 5 years of follow up. RESULTS A total of 508 patients (64% female, 94.9% White, mean age 53.5 ± 10.5 years, BMI (46.0 ± 8.3 kg/m2) were included in the analysis. This study demonstrates a profound association between insulin dosage quartiles and T2D remission after MBS. Patients with lower insulin requirements showed superior remission rates; those in the lowest quartile had remission rates of 73%, 70%, and 62% at 1, 3, and 5 years, respectively, compared to 34%, 37%, and 36% in the highest quartile (P < 0.001 across all intervals). RYGB surgery showed a significantly better remission in the second and third insulin quartiles, suggesting its effectiveness over SG for patients with mid-range insulin requirements. CONCLUSION This study underscores the importance of considering insulin dosage when predicting T2D remission post-MBS. The findings advocate for a more nuanced selection of MBS procedures based on individual insulin profiles, potentially enhancing diabetes remission outcomes.
Collapse
Affiliation(s)
- Wissam Ghusn
- Department of Internal Medicine, Boston University, Boston, MA, USA
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Yara Salameh
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Kamal Abi Mosleh
- Department of Surgery, Division of Metabolic and Abdominal Wall Reconstructive Surgery, Mayo Clinic, Rochester, MN, USA
| | - Meera Shah
- Department of Medicine, Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA
| | - Andrew C Storm
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Barham K Abu Dayyeh
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Omar M Ghanem
- Department of Surgery, Division of Metabolic and Abdominal Wall Reconstructive Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
20
|
Karlsson C, Johnson LK, Greasley PJ, Retterstøl K, Hedberg J, Hall M, Hawker N, Robertsen I, Havsol J, Hertel JK, Sandbu R, Skovlund E, Olsen T, Christensen H, Jansson-Löfmark R, Andersson S, Åsberg A, Hjelmesæth J. Gastric Bypass vs Diet and Cardiovascular Risk Factors: A Nonrandomized Controlled Trial. JAMA Surg 2024; 159:971-980. [PMID: 38959017 PMCID: PMC11223056 DOI: 10.1001/jamasurg.2024.2162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/13/2024] [Indexed: 07/04/2024]
Abstract
Importance Roux-en-Y gastric bypass (RYGB) is associated with reduced cardiovascular (CV) risk factors, morbidity, and mortality. Whether these effects are specifically induced by the surgical procedure or the weight loss is unclear. Objective To compare 6-week changes in CV risk factors in patients with obesity undergoing matching caloric restriction and weight loss by RYGB or a very low-energy diet (VLED). Design, Setting, and Participants This nonrandomized controlled study (Impact of Body Weight, Low Calorie Diet, and Gastric Bypass on Drug Bioavailability, Cardiovascular Risk Factors, and Metabolic Biomarkers [COCKTAIL]) was conducted at a tertiary care obesity center in Norway. Participants were individuals with severe obesity preparing for RYGB or a VLED. Recruitment began February 26, 2015; the first patient visit was on March 18, 2015, and the last patient visit (9-week follow-up) was on August 9, 2017. Data were analyzed from April 30, 2021, through June 29, 2023. Interventions VLED alone for 6 weeks or VLED for 6 weeks after RYGB; both interventions were preceded by 3-week LED. Main Outcomes and Measures Between-group comparisons of 6-week changes in CV risk factors. Results Among 78 patients included in the analyses, the mean (SD) age was 47.5 (9.7) years; 51 (65%) were women, and 27 (35%) were men. Except for a slightly higher mean (SD) body mass index of 44.5 (6.2) in the RYGB group (n = 41) vs 41.9 (5.4) in the VLED group (n = 37), baseline demographic and clinical characteristics were similar between groups. Major atherogenic blood lipids (low-density lipoprotein cholesterol, non-high-density lipoprotein cholesterol, apolipoprotein B, lipoprotein[a]) were reduced after RYGB in comparison with VLED despite a similar fat mass loss. Mean between-group differences were -17.7 mg/dL (95% CI, -27.9 to -7.5), -17.4 mg/dL (95% CI, -29.8 to -5.0) mg/dL, -9.94 mg/dL (95% CI, -15.75 to -4.14), and geometric mean ratio was 0.55 U/L (95% CI, 0.42 to 0.72), respectively. Changes in glycemic control and blood pressure were similar between groups. Conclusions and Relevance This study found that clinically meaningful reductions in major atherogenic blood lipids were demonstrated after RYGB, indicating that RYGB may reduce CV risk independent of weight loss. Trial Registration ClinicalTrials.gov Identifier: NCT02386917.
Collapse
Affiliation(s)
- Cecilia Karlsson
- Late-Stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Line Kristin Johnson
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, Tønsberg, Norway
| | - Peter J. Greasley
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kjetil Retterstøl
- The Lipid Clinic, Oslo University Hospital, Oslo, Norway
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jonatan Hedberg
- Medical Evidence and Observational Research, Global Medical BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Martin Hall
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Noele Hawker
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ida Robertsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Jesper Havsol
- Data Science and Artificial Intelligence, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jens Kristoffer Hertel
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, Tønsberg, Norway
| | - Rune Sandbu
- Department of Surgery, Vestfold Hospital Trust, Tønsberg, Norway
| | - Eva Skovlund
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hege Christensen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Rasmus Jansson-Löfmark
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Shalini Andersson
- Research and Early Development, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anders Åsberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
- Department of Transplantation Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Jøran Hjelmesæth
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, Tønsberg, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
21
|
Taylor R. Understanding the cause of type 2 diabetes. Lancet Diabetes Endocrinol 2024; 12:664-673. [PMID: 39038473 DOI: 10.1016/s2213-8587(24)00157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/24/2024]
Abstract
Type 2 diabetes has long been thought to have heterogenous causes, even though epidemiological studies uniformly show a tight relationship with overnutrition. The twin cycle hypothesis postulated that interaction of self-reinforcing cycles of fat accumulation inside the liver and pancreas, driven by modest but chronic positive calorie balance, could explain the development of type 2 diabetes. This hypothesis predicted that substantial weight loss would bring about a return to the non-diabetic state, permitting observation of the pathophysiology determining the transition. These changes were postulated to reflect the basic mechanisms of causation in reverse. A series of studies over the past 15 years has elucidated these underlying mechanisms. Together with other research, the interaction of environmental and genetic factors has been clarified. This knowledge has led to successful implementation of a national programme for remission of type 2 diabetes. This Review discusses the paucity of evidence for heterogeneity in causes of type 2 diabetes and summarises the in vivo pathophysiological changes, which cause this disease of overnutrition. Type 2 diabetes has a homogenous cause expressed in genetically heterogenous individuals.
Collapse
Affiliation(s)
- Roy Taylor
- Newcastle Magnetic Resonance Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
22
|
Seo E, Kwon Y, ALRomi A, Eledreesi M, Park S. A multifaceted and inclusive methodology for the detection of sarcopenia in patients undergoing bariatric surgery: an in-depth analysis of current evidence. Rev Endocr Metab Disord 2024; 25:677-689. [PMID: 38427134 DOI: 10.1007/s11154-023-09864-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 03/02/2024]
Abstract
Bariatric surgery is widely recognized as the most effective intervention for obesity and offers benefits beyond weight loss. However, not all patients achieve satisfactory weight loss, balanced changes in body composition, and resolution of comorbidities. Therefore, thorough pre- and postoperative evaluations are important to predict success and minimize adverse effects. More comprehensive assessments require broadening the focus beyond body weight and fat measurements to consider quantitative and qualitative evaluations of muscles. Introducing the concept of sarcopenia is useful for assessing the degradative and pathological changes in muscles associated with cardiometabolic function, physical performance, and other obesity-related comorbidities in patients undergoing bariatric surgery. However, there is currently no consensus or definition regarding the research and clinical use of sarcopenia in patients undergoing bariatric surgery. Therefore, this review aimed to define the concept of sarcopenia applicable to patients undergoing bariatric surgery, based on the consensus reached for sarcopenia in the general population. We also discuss the methods and significance of measuring muscle mass, quality, and strength, which are key variables requiring a comprehensive assessment.
Collapse
Affiliation(s)
- Eunhye Seo
- Keimyung University College of Nursing, Daegu, Republic of Korea
| | - Yeongkeun Kwon
- Division of Foregut Surgery, Korea University College of Medicine, 73 Goryeodae- ro, Seongbuk-gu, 02841, Seoul, Republic of Korea
- Center for Obesity and Metabolic Diseases, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Ahmad ALRomi
- Ministry of Health Jordan, General surgery affiliationision, prince Hamzah hospital, Amman, Jordan
| | | | - Sungsoo Park
- Division of Foregut Surgery, Korea University College of Medicine, 73 Goryeodae- ro, Seongbuk-gu, 02841, Seoul, Republic of Korea.
- Center for Obesity and Metabolic Diseases, Korea University Anam Hospital, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Huttasch M, Roden M, Kahl S. Obesity and MASLD: Is weight loss the (only) key to treat metabolic liver disease? Metabolism 2024; 157:155937. [PMID: 38782182 DOI: 10.1016/j.metabol.2024.155937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 04/25/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) closely associates with obesity and type 2 diabetes. Lifestyle intervention and bariatric surgery aiming at substantial weight loss are cornerstones of MASLD treatment by improving histological outcomes and reducing risks of comorbidities. Originally developed as antihyperglycemic drugs, incretin (co-)agonists and SGLT2 inhibitors also reduce steatosis and cardiorenovascular events. Certain incretin agonists effectively improve histological features of MASLD, but not fibrosis. Of note, beneficial effects on MASLD may not necessarily require weight loss. Despite moderate weight gain, one PPARγ agonist improved adipose tissue and MASLD with certain benefit on fibrosis in post-hoc analyses. Likewise, the first THRβ-agonist was recently provisionally approved because of significant improvements of MASLD and fibrosis. We here discuss liver-related and metabolic effects induced by different MASLD treatments and their association with weight loss. Therefore, we compare results from clinical trials on drugs acting via weight loss (incretin (co)agonists, SGLT2 inhibitors) with those exerting no weight loss (pioglitazone; resmetirom). Furthermore, other drugs in development directly targeting hepatic lipid metabolism (lipogenesis inhibitors, FGF21 analogs) are addressed. Although THRβ-agonism may effectively improve hepatic outcomes, MASLD treatment concepts should consider all cardiometabolic risk factors for effective reduction of morbidity and mortality in the affected people.
Collapse
Affiliation(s)
- Maximilian Huttasch
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany.
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany; Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Sabine Kahl
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine University Düsseldorf, Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany.
| |
Collapse
|
24
|
Wahlström A, Aydin Ö, Olsson LM, Sjöland W, Henricsson M, Lundqvist A, Marschall HU, Franken R, van de Laar A, Gerdes V, Meijnikman AS, Hofsø D, Groen AK, Hjelmesæth J, Nieuwdorp M, Bäckhed F. Alterations in bile acid kinetics after bariatric surgery in patients with obesity with or without type 2 diabetes. EBioMedicine 2024; 106:105265. [PMID: 39096744 PMCID: PMC11345581 DOI: 10.1016/j.ebiom.2024.105265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/17/2024] [Accepted: 07/21/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND Bariatric surgery is an effective treatment option for obesity and provides long-term weight loss and positive effects on metabolism, but the underlying mechanisms are poorly understood. Alterations in bile acid metabolism have been suggested as a potential contributing factor, but comprehensive studies in humans are lacking. METHODS In this study, we analysed the postprandial responses of bile acids, C4 and FGF19 in plasma, and excretion of bile acids in faeces, before and after bariatric surgery in patients (n = 38; 74% females) with obesity with or without type 2 diabetes from the BARIA cohort. FINDINGS We observed that total fasting plasma bile acid levels increased, and faecal excretion of bile acids decreased after surgery suggesting increased reabsorption of bile acids. Consistent with increased bile acid levels after surgery we observed increased postprandial levels of FGF19 and suppression of the bile acid synthesis marker C4, suggesting increased FXR activation in the gut. We also noted that a subset of bile acids had altered postprandial responses before and after surgery. Finally, fasting plasma levels of 6α-hydroxylated bile acids, which are TGR5 agonists and associated with improved glucose metabolism, were increased after surgery and one of them, HDCA, covaried with diabetes remission in an independent cohort. INTERPRETATION Our findings provide new insights regarding bile acid kinetics and suggest that bariatric surgery in humans alters bile acid profiles leading to activation of FXR and TGR5, which may contribute to weight loss, improvements in glucose metabolism, and diabetes remission. FUNDING Novo Nordisk Fonden, Leducq Foundation, Swedish Heart-Lung Foundation, Knut and Alice Wallenberg Foundation, the ALF-agreement, ZonMw.
Collapse
Affiliation(s)
- Annika Wahlström
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg S-413 45, Sweden
| | - Ömrüm Aydin
- Department of Internal and Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands; Department of Internal Medicine, Spaarne Gasthuis, Hoofddorp, the Netherlands
| | - Lisa M Olsson
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg S-413 45, Sweden
| | - Wilhelm Sjöland
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg S-413 45, Sweden
| | - Marcus Henricsson
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg S-413 45, Sweden
| | - Annika Lundqvist
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg S-413 45, Sweden
| | - Hanns-Ulrich Marschall
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg S-413 45, Sweden
| | - Rutger Franken
- Department of Surgery, Spaarne Hospital, Hoofddorp, the Netherlands
| | | | - Victor Gerdes
- Department of Internal and Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Abraham S Meijnikman
- Department of Internal and Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Dag Hofsø
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, Tønsberg, Norway
| | - Albert K Groen
- Experimental Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jøran Hjelmesæth
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, Tønsberg, Norway; Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, Norway
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands; Experimental Vascular Medicine, Amsterdam UMC, Amsterdam, the Netherlands
| | - Fredrik Bäckhed
- Wallenberg Laboratory and Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg S-413 45, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark; Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden.
| |
Collapse
|
25
|
Sun CJ, Nguyen É, Prentki M, Rabasa-Lhoret R. The daunting task of recruiting insulin-treated patients living with type 2 diabetes mellitus for intensive lifestyle interventions. Diabetes Obes Metab 2024; 26:3486-3488. [PMID: 38840460 DOI: 10.1111/dom.15688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Affiliation(s)
- Cathy J Sun
- Division of Endocrinology and Metabolism, Department of Medicine, The Ottawa Hospital, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Montreal Clinical Research Institute, Montreal, Québec, Canada
| | | | - Marc Prentki
- Departments of Nutrition, Biochemistry and Molecular Medicine, Université de Montréal, Montreal Diabetes Research Center, CRCHUM, Montreal, Québec, Canada
- Division of Endocrinology and Metabolism, Department of Medicine and Nutrition, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Rémi Rabasa-Lhoret
- Montreal Clinical Research Institute, Montreal, Québec, Canada
- Division of Endocrinology and Metabolism, Department of Medicine and Nutrition, Faculty of Medicine, Université de Montréal, Montreal, Québec, Canada
| |
Collapse
|
26
|
Kajani S, Laker RC, Ratkova E, Will S, Rhodes CJ. Hepatic glucagon action: beyond glucose mobilization. Physiol Rev 2024; 104:1021-1060. [PMID: 38300523 DOI: 10.1152/physrev.00028.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Glucagon's ability to promote hepatic glucose production has been known for over a century, with initial observations touting this hormone as a diabetogenic agent. However, glucagon receptor agonism [when balanced with an incretin, including glucagon-like peptide 1 (GLP-1) to dampen glucose excursions] is now being developed as a promising therapeutic target in the treatment of metabolic diseases, like metabolic dysfunction-associated steatotic disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH), and may also have benefit for obesity and chronic kidney disease. Conventionally regarded as the opposing tag-team partner of the anabolic mediator insulin, glucagon is gradually emerging as more than just a "catabolic hormone." Glucagon action on glucose homeostasis within the liver has been well characterized. However, growing evidence, in part thanks to new and sensitive "omics" technologies, has implicated glucagon as more than just a "glucose liberator." Elucidation of glucagon's capacity to increase fatty acid oxidation while attenuating endogenous lipid synthesis speaks to the dichotomous nature of the hormone. Furthermore, glucagon action is not limited to just glucose homeostasis and lipid metabolism, as traditionally reported. Glucagon plays key regulatory roles in hepatic amino acid and ketone body metabolism, as well as mitochondrial turnover and function, indicating broader glucagon signaling consequences for metabolic homeostasis mediated by the liver. Here we examine the broadening role of glucagon signaling within the hepatocyte and question the current dogma, to appreciate glucagon as more than just that "catabolic hormone."
Collapse
Affiliation(s)
- Sarina Kajani
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Rhianna C Laker
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Ekaterina Ratkova
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sarah Will
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Christopher J Rhodes
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| |
Collapse
|
27
|
Morissette A, Mulvihill EE. Obesity management for the treatment of type 2 diabetes: emerging evidence and therapeutic approaches. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13065. [PMID: 38903652 PMCID: PMC11186996 DOI: 10.3389/jpps.2024.13065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
Excess adiposity can contribute to metabolic complications, such as type 2 diabetes mellitus (T2DM), which poses a significant global health burden. Traditionally viewed as a chronic and irreversible condition, T2DM management has evolved and new approaches emphasizing reversal and remission are emerging. Bariatric surgery demonstrates significant improvements in body weight and glucose homeostasis. However, its complexity limits widespread implementation as a population-wide intervention. The identification of glucagon-like peptide 1 (GLP-1) and the development of GLP-1 receptor agonists (GLP-1RAs) have improved T2DM management and offer promising outcomes in terms of weight loss. Innovative treatment approaches combining GLP-1RA with other gut and pancreatic-derived hormone receptor agonists, such as glucose-dependant insulinotropic peptide (GIP) and glucagon (GCG) receptor agonists, or coadministered with amylin analogues, are demonstrating enhanced efficacy in both weight loss and glycemic control. This review aims to explore the benefits of bariatric surgery and emerging pharmacological therapies such as GLP-1RAs, and dual and triple agonists in managing obesity and T2DM while highlighting the caveats and evolving landscape of treatment options.
Collapse
Affiliation(s)
| | - Erin E. Mulvihill
- The University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Faculty of Medicine, Ottawa, ON, Canada
| |
Collapse
|
28
|
Ghanem OM, Abi Mosleh K, Kerbage A, Lu L, Hage K, Abu Dayyeh BK. Continued Diabetes Remission Despite Weight Recurrence: Gastric Bypass Long-Term Metabolic Benefit. J Am Coll Surg 2024; 238:862-871. [PMID: 38349010 DOI: 10.1097/xcs.0000000000000934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) demonstrates high rates of type 2 diabetes mellitus (T2DM) remission, a phenomenon hypothesized to be mediated mainly by weight loss. Compared with procedures that do not bypass the proximal small intestines, such as sleeve gastrectomy (SG), RYGB exhibits weight loss-independent intestinal mechanisms conducive to T2DM remission. We investigated continued diabetes remission (CDR) rates despite weight recurrence (WR) after RYGB compared with an SG cohort. STUDY DESIGN A retrospective review of patients who underwent successful primary RYGB or SG with a BMI value of 35 kg/m 2 or more and a preoperative diagnosis of T2DM was performed. Patients with less than 5 years of follow-up, absence of WR, or lack of T2DM remission at nadir weight were excluded. After selecting the optimal procedure for glycemic control, rates of CDR were then stratified into WR quartiles and compared. RESULTS A total of 224 RYGB and 46 SG patients were analyzed. The overall rate of CDR was significantly higher in the RYGB group (75%) compared with the SG group (34.8%; p < 0.001). The odds of T2DM recurrence were 5.5 times higher after SG compared with RYGB. Rates of CDR were stratified into WR quartiles (85.5%, <25%; 81.7%, 25% to 44.9%; 63.2%, 45% to 74.9%; and 60%, >75%). Baseline insulin use, higher preoperative glycosylated hemoglobin, and longer preoperative duration of T2DM were associated with T2DM recurrence, whereas WR was not. CONCLUSIONS T2DM remission rates after RYGB are maintained despite WR, arguing for a concurrent weight loss-independent metabolic benefit likely facilitated by bypassing the proximal small intestine.
Collapse
Affiliation(s)
- Omar M Ghanem
- From the Department of Surgery (Ghanem, Abi Mosleh, Hage), Mayo Clinic, Rochester, MN
| | - Kamal Abi Mosleh
- From the Department of Surgery (Ghanem, Abi Mosleh, Hage), Mayo Clinic, Rochester, MN
| | - Anthony Kerbage
- Division of Gastroenterology, Department of Medicine (Kerbage, Abu Dayyeh), Mayo Clinic, Rochester, MN
| | - Lauren Lu
- Mayo Clinic Alix School of Medicine (Lu), Mayo Clinic, Rochester, MN
| | - Karl Hage
- From the Department of Surgery (Ghanem, Abi Mosleh, Hage), Mayo Clinic, Rochester, MN
| | - Barham K Abu Dayyeh
- Division of Gastroenterology, Department of Medicine (Kerbage, Abu Dayyeh), Mayo Clinic, Rochester, MN
| |
Collapse
|
29
|
Zhao Z, Liu S, Qian B, Zhou L, Shi J, Liu J, Xu L, Yang Z. CMKLR1 senses chemerin/resolvin E1 to control adipose thermogenesis and modulate metabolic homeostasis. FUNDAMENTAL RESEARCH 2024; 4:575-588. [PMID: 38933207 PMCID: PMC11197767 DOI: 10.1016/j.fmre.2022.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/01/2022] [Accepted: 06/22/2022] [Indexed: 11/18/2022] Open
Abstract
Induction of beige fat for thermogenesis is a potential therapy to improve homeostasis against obesity. β3-adrenoceptor (β3-AR), a type of G protein-coupled receptor (GPCR), is believed to mediate the thermogenesis of brown fat in mice. However, β3-AR has low expression in human adipose tissue, precluding its activation as a standalone clinical modality. This study aimed at identifying a potential GPCR target to induce beige fat. We found that chemerin chemokine-like receptor 1 (CMKLR1), one of the novel GPCRs, mediated the development of beige fat via its two ligands, chemerin and resolvin E1 (RvE1). The RvE1 levels were decreased in the obese mice, and RvE1 treatment led to a substantial improvement in obese features and augmented beige fat markers. Inversely, despite sharing the same receptor as RvE1, the chemerin levels were increased in obesogenic conditions, and chemerin treatment led to an augmented obese phenotype and a decline of beige fat markers. Moreover, RvE1 and chemerin induced or restrained the development of beige fat, respectively, via the mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway. We further showed that RvE1 and chemerin regulated mTORC1 signaling differentially by forming hydrogen bonds with different binding sites of CMKLR1. In conclusion, our study showed that RvE1 and chemerin affected metabolic homeostasis differentially, suggesting that selectively modulating CMKLR1 may be a potential therapeutic target for restoring metabolic homeostasis.
Collapse
Affiliation(s)
- Zewei Zhao
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University; Shenzhen, Guangdong 518107, China
| | - Siqi Liu
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University; Shenzhen, Guangdong 518107, China
| | - Bingxiu Qian
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University; Shenzhen, Guangdong 518107, China
| | - Lin Zhou
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University; Guangzhou, Guangdong 510080, China
| | - Jianglin Shi
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University; Shenzhen, Guangdong 518107, China
| | - Junxi Liu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University; Guangzhou, Guangdong 510080, China
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Zhonghan Yang
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University; Shenzhen, Guangdong 518107, China
| |
Collapse
|
30
|
Retnakaran R, Kashyap SR, Gerstein HC, Aroda VR. Contemporary Clinical Perspectives on Targeting Remission of Type 2 Diabetes. J Clin Endocrinol Metab 2024; 109:1179-1188. [PMID: 38108415 DOI: 10.1210/clinem/dgad746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 12/19/2023]
Abstract
It has long been known that some patients with type 2 diabetes (T2DM) can experience sustained metabolic improvement to near-normal levels of glycemia either spontaneously or after medical intervention. Now recognized as remission of diabetes, this intriguing state is currently more feasible than ever before due to profound advances in metabolic surgery, pharmacologic therapy, and regimens of lifestyle modification. This enhanced capacity to induce remission has revealed new pathophysiologic insights, including the presence of a reversible component of the pancreatic beta-cell dysfunction that otherwise drives the chronic progressive nature of T2DM. In doing so, it has changed the therapeutic landscape by offering new potential management objectives and considerations for patients and providers. However, the excitement around these developments must also be tempered by the sobering realities of our current understanding of remission, including the recognition that this condition may not be permanent (resulting in glycemic relapse over time) and that beta-cell function may not be normalized in the setting of remission. These limitations highlight both the many gaps in our current understanding of remission and the caution with which clinical discussions must be handled for clear patient-directed communication of the pros and cons of targeting this outcome in practice. In this mini-review, we consider this rapidly growing literature, including its implications and its limitations, and thereby seek to provide objective balanced perspectives on targeting remission of T2DM in current clinical care.
Collapse
Affiliation(s)
- Ravi Retnakaran
- Leadership Sinai Centre for Diabetes, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada
- Division of Endocrinology, University of Toronto, Toronto, ON M5S 3H2, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Sangeeta R Kashyap
- Division of Endocrinology, Diabetes, and Metabolism, New York Presbyterian-Weill Cornell Medicine, New York, NY 10021, USA
| | - Hertzel C Gerstein
- Division of Endocrinology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Vanita R Aroda
- Division of Endocrinology, Diabetes & Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
31
|
Lannoo M, Simoens C, Vangoitsenhoven R, Gillard P, D'Hoore A, De Vadder M, Mertens A, Deleus E, Steenackers N, Mathieu C, Van der Schueren B. Comparative impact of Roux-en-Y gastric bypass, sleeve gastrectomy or diet alone on beta-cell function in insulin-treated type 2 diabetes patients. Sci Rep 2024; 14:8211. [PMID: 38589596 PMCID: PMC11001928 DOI: 10.1038/s41598-024-59048-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/05/2024] [Indexed: 04/10/2024] Open
Abstract
Although bariatric surgery is an effective treatment for type 2 diabetes by inducing weight loss and augmenting gut hormone secretion, the immediate effect on beta-cell function itself remains to be elucidated in type 2 diabetes. Therefore, a prospective, randomized trial was performed in 30 patients with insulin-treated type 2 diabetes and a body mass index ≥ 35 kg/m2. Patients were randomly assigned (1:1:1) to Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG) in combination with protein-sparing modified fast (PSMF), or to PSMF alone. Eu- and hyperglycemic clamps were performed before and 3 weeks after surgery and/or PSMF initiation. The primary outcome was the evolution of insulin sensitivity and beta-cell function after surgery, calculated using the composite measures of glucose disposal rate, insulin secretion rate, and disposition index (DI). Results revealed that markers of insulin sensitivity increased similarly in all arms (p = 0.43). A higher marker for maximal beta-cell function was observed when comparing SG to PSMF (p = 0.007). The DI showed a clear positive evolution after RYGB and SG, but not after PSMF alone. Altogether, these findings indicate that bariatric surgery results in an immediate beta-cell function recovery in insulin-treated type 2 diabetes.
Collapse
Affiliation(s)
- Matthias Lannoo
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Caroline Simoens
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Roman Vangoitsenhoven
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Pieter Gillard
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - André D'Hoore
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Mieke De Vadder
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Ann Mertens
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Ellen Deleus
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Nele Steenackers
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Bart Van der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium.
- Department of Endocrinology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
32
|
Stevenson M, Srivastava A, Nacher M, Hall C, Palaia T, Lee J, Zhao CL, Lau R, Ali MAE, Park CY, Schlamp F, Heffron SP, Fisher EA, Brathwaite C, Ragolia L. The Effect of Diet Composition on the Post-operative Outcomes of Roux-en-Y Gastric Bypass in Mice. Obes Surg 2024; 34:911-927. [PMID: 38191966 PMCID: PMC11926884 DOI: 10.1007/s11695-023-07052-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/30/2023] [Accepted: 12/30/2023] [Indexed: 01/10/2024]
Abstract
PURPOSE Roux-en-Y gastric bypass (RYGB) leads to the improvement of many obesity-associated conditions. The degree to which post-operative macronutrient composition contributes to metabolic improvement after RYGB is understudied. METHODS A mouse model of RYGB was used to examine the effects of diet on the post-operative outcomes of RYGB. Obese mice underwent either Sham or RYGB surgery and were administered either chow or HFD and then monitored for an additional 8 weeks. RESULTS After RYGB, reductions to body weight, fat mass, and lean mass were similar regardless of diet. RYGB and HFD were independently detrimental to bone mineral density and plasma vitamin D levels. Independent of surgery, HFD accelerated hematopoietic stem and progenitor cell proliferation and differentiation and exhibited greater myeloid lineage commitment. Independent of diet, systemic iron deficiency was present after RYGB. In both Sham and RYGB groups, HFD increased energy expenditure. RYGB increased fecal energy loss, and HFD after RYGB increased fecal lipid content. RYGB lowered fasting glucose and liver glycogen levels but HFD had an opposing effect. Indices of insulin sensitivity improved independent of diet. HFD impaired improvements to dyslipidemia, NAFLD, and fibrosis. CONCLUSION Post-operative diet plays a significant role in determining the degree to which RYGB reverses obesity-induced metabolic abnormalities such as hyperglycemia, dyslipidemia, and NAFLD. Diet composition may be targeted in order to assist in the treatment of post-RYGB bone mineral density loss and vitamin D deficiency as well as to reverse myeloid lineage commitment. HFD after RYGB continues to pose a significant multidimensional health risk.
Collapse
Affiliation(s)
- Matthew Stevenson
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Ankita Srivastava
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Maria Nacher
- Department of Medicine, Division of Cardiology, NYU Langone Health Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology and the Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Christopher Hall
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Thomas Palaia
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Jenny Lee
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Chaohui Lisa Zhao
- Department of Pathology, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Raymond Lau
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
- Department of Endocrinology, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Mohamed A E Ali
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Christopher Y Park
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Florencia Schlamp
- Department of Medicine, Division of Cardiology, NYU Langone Health Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Sean P Heffron
- Department of Medicine, Division of Cardiology, NYU Langone Health Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology and the Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, NYU Langone Health Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology and the Cardiovascular Research Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Collin Brathwaite
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA
- Department of Surgery, NYU Langone Hospital-Long Island, Mineola, NY, USA
| | - Louis Ragolia
- Department of Biomedical Research, NYU Grossman Long Island School of Medicine, NYU Langone Hospital-Long Island, Mineola, NY, USA.
- Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
33
|
Cao C, Koh HCE, Reeds DN, Patterson BW, Klein S, Mittendorfer B. Critical Evaluation of Indices Used to Assess β-Cell Function. Diabetes 2024; 73:391-400. [PMID: 38015795 PMCID: PMC10882145 DOI: 10.2337/db23-0613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/18/2023] [Indexed: 11/30/2023]
Abstract
The assessment of β-cell function, defined as the relationship between insulin secretion rate (ISR) and plasma glucose, is not standardized and often involves any of a number of β-cell function indices. We compared β-cell function by using popular indices obtained during basal conditions and after glucose ingestion, including the HOMA-B index, the basal ISR (or plasma insulin)-to-plasma glucose concentration ratio, the insulinogenic and ISRogenic indices, the ISR (or plasma insulin)-to-plasma glucose concentration areas (or incremental areas) under the curve ratio, and the disposition index, which integrates a specific β-cell function index value with an estimate of insulin sensitivity, between lean people with normal fasting glucose (NFG) and normal glucose tolerance (NGT) (n = 50) and four groups of people with obesity (n = 188) with 1) NFG-NGT, 2) NFG and impaired glucose tolerance (IGT), 3) impaired fasting glucose (IFG) and IGT, and 4) type 2 diabetes. We also plotted the ISR-plasma glucose relationship before and after glucose ingestion and used a statistical mixed-effects model to evaluate group differences in this relationship (i.e., β-cell function). Index-based group differences in β-cell function produced contradicting results and did not reflect the group differences of the actual observed ISR-glucose relationship or, in the case of the disposition index, group differences in glycemic status. The discrepancy in results is likely due to incorrect mathematical assumptions that are involved in computing indices, which can be overcome by evaluating the relationship between ISR and plasma glucose with an appropriate statistical model. Data obtained with common β-cell function indices should be interpreted cautiously. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Chao Cao
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Han-Chow E. Koh
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Dominic N. Reeds
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Bruce W. Patterson
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
- Sansum Diabetes Research Institute, Santa Barbara, CA
| | - Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO
- Departments of Medicine and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| |
Collapse
|
34
|
Mittendorfer B, van Vliet S, Smith GI, Petersen MC, Patterson BW, Klein S. Impaired plasma glucose clearance is a key determinant of fasting hyperglycemia in people with obesity. Obesity (Silver Spring) 2024; 32:540-546. [PMID: 38228469 PMCID: PMC10922622 DOI: 10.1002/oby.23963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 01/18/2024]
Abstract
OBJECTIVE The objective of this study was to evaluate the relative importance of the basal rate of glucose appearance (Ra) in the circulation and the basal rate of plasma glucose clearance in determining fasting plasma glucose concentration in people with obesity and different fasting glycemic statuses. METHODS The authors evaluated basal glucose kinetics in 33 lean people with normal fasting glucose (<100 mg/dL; Lean < 100 group) and 206 people with obesity and normal fasting glucose (Ob < 100 group, n = 118), impaired fasting glucose (100-125 mg/dL; Ob 100-125 group, n = 66), or fasting glucose diagnostic of diabetes (≥126 mg/dL; Ob ≥ 126 group, n = 22). RESULTS Although there was a large (up to three-fold) range in glucose Ra within each group, the ranges in glucose concentration in the Lean < 100, Ob < 100, and Ob 100-125 groups were small because of a close relationship between glucose Ra and clearance rate. However, the glucose clearance rate at any Ra value was lower in the hyperglycemic than the normoglycemic groups. In the Ob ≥ 126 group, plasma glucose concentration was primarily determined by glucose Ra, because glucose clearance was markedly attenuated. CONCLUSIONS Fasting hyperglycemia in people with obesity represents a disruption of the precisely regulated integration of glucose production and clearance rates.
Collapse
Affiliation(s)
- Bettina Mittendorfer
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
- University of Missouri, School of Medicine in Columbia, MO, USA
| | - Stephan van Vliet
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Gordon I. Smith
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Max C. Petersen
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruce W. Patterson
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Klein
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO, USA
- Sansum Diabetes Research Institute, Santa Barbara, CA, USA
| |
Collapse
|
35
|
Onishi Y, Oura T, Takeuchi M. Metabolic Abnormalities Following Tirzepatide Monotherapy in Japanese Patients with Type 2 Diabetes: A Phase 3 SURPASS J-mono Post Hoc Analysis. Diabetes Ther 2024; 15:649-661. [PMID: 38310163 PMCID: PMC10942919 DOI: 10.1007/s13300-024-01534-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 01/15/2024] [Indexed: 02/05/2024] Open
Abstract
INTRODUCTION The presence of metabolic abnormalities in patients with type 2 diabetes (T2D) increases the risk of cardiovascular disease and other comorbidities. This analysis compared the effects of tirzepatide (5, 10, and 15 mg) and dulaglutide 0.75 mg on the prevalence of metabolic abnormalities in Japanese patients with T2D. METHODS This was a post hoc analysis of SURPASS J-mono, a multicenter, randomized, double-blind, active-controlled, parallel-group, phase 3 trial that compared the efficacy and safety of tirzepatide monotherapy (5, 10, and 15 mg) to dulaglutide 0.75 mg in Japanese patients with T2D. Thresholds for abnormalities were based on the Japanese criteria for metabolic syndrome. Proportions of participants meeting a composite endpoint (visceral fat accumulation measured by waist circumference plus two or more of dyslipidemia, hypertension, or hyperglycemia) or individual component thresholds were calculated at baseline and week 52 for the overall population and for baseline body mass index (BMI) subgroups (< 25, 25 to < 30, and ≥ 30 kg/m2). RESULTS Of 636 randomized participants, 431 (67.8%) met the composite endpoint at baseline, with similar findings observed across treatment arms. At week 52, the proportion of participants on treatment that met the composite endpoint was 31.7%, 23.0%, and 14.2% in the tirzepatide 5-, 10-, and 15-mg arms, respectively, and 56.5% in the dulaglutide arm (p < 0.001). A higher proportion met the composite endpoint at baseline in the BMI 25 to < 30 and ≥ 30 kg/m2 subgroups (73.2-79.3%) compared with the < 25 kg/m2 subgroup (45.3%), with reductions observed across all BMI subgroups treated with tirzepatide. The proportion of participants with individual metabolic abnormalities showed similar trends to those observed for the composite endpoint. Tirzepatide was consistently superior to dulaglutide across all assessments. CONCLUSIONS Tirzepatide reduced the prevalence of multiple metabolic abnormalities, indicating tirzepatide may have metabolic benefit in Japanese patients with T2D. TRIAL REGISTRATION ClinicalTrials.gov, NCT03861052.
Collapse
Affiliation(s)
- Yukiko Onishi
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo, Japan
| | - Tomonori Oura
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K, Kobe, 651-0086, Japan
| | - Masakazu Takeuchi
- Japan Drug Development and Medical Affairs, Eli Lilly Japan K.K, Kobe, 651-0086, Japan.
| |
Collapse
|
36
|
Bloomgarden Z. The WCIRDC 2023: Concepts of insulin resistance. J Diabetes 2024; 16:e13552. [PMID: 38516866 PMCID: PMC10958406 DOI: 10.1111/1753-0407.13552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/06/2024] [Indexed: 03/23/2024] Open
|
37
|
Drucker DJ. Prevention of cardiorenal complications in people with type 2 diabetes and obesity. Cell Metab 2024; 36:338-353. [PMID: 38198966 DOI: 10.1016/j.cmet.2023.12.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024]
Abstract
Traditional approaches to prevention of the complications of type 2 diabetes (T2D) and obesity have focused on reduction of blood glucose and body weight. The development of new classes of medications, together with evidence from dietary weight loss and bariatric surgery trials, provides new options for prevention of heart failure, chronic kidney disease, myocardial infarction, stroke, metabolic liver disease, cancer, T2D, and neurodegenerative disorders. Here I review evidence for use of lifestyle modification, SGLT-2 inhibitors, GLP-1 receptor agonists, selective mineralocorticoid receptor antagonists, and bariatric surgery, for prevention of cardiorenal and metabolic complications in people with T2D or obesity, highlighting the contributions of weight loss, as well as weight loss-independent mechanisms of action. Collectively, the evidence supports a tailored approach to selection of therapeutic interventions for T2D and obesity based on the likelihood of developing specific complications, rather than a stepwise approach focused exclusively on glycemic or weight control.
Collapse
Affiliation(s)
- Daniel Joshua Drucker
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, ON M5G1X5, Canada.
| |
Collapse
|
38
|
Becerril S, Cienfuegos JA, Rodríguez A, Catalán V, Ramírez B, Valentí V, Moncada R, Unamuno X, Gómez-Ambrosi J, Frühbeck G. Single anastomosis duodeno-ileal bypass with sleeve gastrectomy generates sustained improvement of glycemic control compared with sleeve gastrectomy in the diet-induced obese rat model. J Physiol Biochem 2024; 80:149-160. [PMID: 37935948 PMCID: PMC10810039 DOI: 10.1007/s13105-023-00993-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023]
Abstract
Bariatric surgery has become a recognized and effective procedure for treating obesity and type 2 diabetes (T2D). Our objective was to directly compare the caloric intake-independent effects of sleeve gastrectomy (SG) and single anastomosis duodenoileal bypass with SG (SADI-S) on glucose tolerance in rats with diet-induced obesity (DIO) and to elucidate the differences between bariatric surgery and caloric restriction.A total of 120 adult male Wistar rats with DIO and insulin resistance were randomly assigned to surgical (sham operation, SG, and SADI-S) and dietary (pair-feeding the amount of food eaten by animals undergoing the SG or SADI-S surgeries) interventions. Body weight and food intake were weekly monitored, and 6 weeks after interventions, fasting plasma glucose, oral glucose and insulin tolerance tests, plasma insulin, adiponectin, GIP, GLP-1, and ghrelin levels were determined.The body weight of SADI-S rats was significantly (p < 0.001) lower as compared to the sham-operated, SG, and pair-fed groups. Furthermore, SADI-S rats exhibited decreased whole body fat mass (p < 0.001), lower food efficiency rates (p < 0.001), and increased insulin sensitivity, as well as improved glucose and lipid metabolism compared to that of the SG and pair-fed rats.SADI-S was more effective than SG, or caloric restriction, in improving glycemic control and metabolic profile, with a higher remission of insulin resistance as well as long-term weight loss.
Collapse
Affiliation(s)
- Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.
| | - Javier A Cienfuegos
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, 31008, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, 31008, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Xabier Unamuno
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Medical Engineering Laboratory, University of Navarra, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Avda. Pío XII, 36, 31008, Pamplona, Spain.
- CIBER Fisiopatología de La Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain.
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain.
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Avda. Pío XII, 36, Pamplona, Spain.
| |
Collapse
|
39
|
ElSayed NA, Aleppo G, Bannuru RR, Bruemmer D, Collins BS, Ekhlaspour L, Gaglia JL, Hilliard ME, Johnson EL, Khunti K, Lingvay I, Matfin G, McCoy RG, Perry ML, Pilla SJ, Polsky S, Prahalad P, Pratley RE, Segal AR, Seley JJ, Selvin E, Stanton RC, Gabbay RA. 2. Diagnosis and Classification of Diabetes: Standards of Care in Diabetes-2024. Diabetes Care 2024; 47:S20-S42. [PMID: 38078589 PMCID: PMC10725812 DOI: 10.2337/dc24-s002] [Citation(s) in RCA: 440] [Impact Index Per Article: 440.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The American Diabetes Association (ADA) "Standards of Care in Diabetes" includes the ADA's current clinical practice recommendations and is intended to provide the components of diabetes care, general treatment goals and guidelines, and tools to evaluate quality of care. Members of the ADA Professional Practice Committee, an interprofessional expert committee, are responsible for updating the Standards of Care annually, or more frequently as warranted. For a detailed description of ADA standards, statements, and reports, as well as the evidence-grading system for ADA's clinical practice recommendations and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.
Collapse
|
40
|
Wu H, Yang A, Lau ESH, Zhang X, Fan B, Ma RCW, Kong APS, Chow E, So WY, Chan JCN, Luk AOY. 1-year weight change after diabetes diagnosis and long-term incidence and sustainability of remission of type 2 diabetes in real-world settings in Hong Kong: An observational cohort study. PLoS Med 2024; 21:e1004327. [PMID: 38261560 PMCID: PMC10805283 DOI: 10.1371/journal.pmed.1004327] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/05/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Clinical trials have demonstrated that remission of type 2 diabetes can be achieved following sustained weight loss. However, the feasibility of achieving diabetes remission through weight management in real-world settings remains unclear. In this study, we aimed to examine the association of weight change at 1 year after diabetes diagnosis with long-term incidence and sustainability of type 2 diabetes remission in real-world settings in Hong Kong. METHODS AND FINDINGS This was a population-based observational cohort study. The territory-wide Risk Assessment and Management Programme for Diabetes Mellitus (RAMP-DM) provides regular comprehensive assessments of metabolic control and complication screening for people with diabetes in Hong Kong. We included 37,326 people with newly diagnosed type 2 diabetes who were enrolled in the RAMP-DM between 2000 and 2017, followed until 2019. Diabetes remission was defined as 2 consecutive HbA1c <6.5% measurements at least 6 months apart in the absence of glucose-lowering drugs (GLDs) and with no record of GLDs at least 3 months before these measurements. During a median follow-up of 7.9 years, 6.1% (2,279) of people achieved diabetes remission, with an incidence rate of 7.8 (95% CI: 7.5, 8.1) per 1,000 person-years. After adjusting for age at diabetes diagnosis, sex, assessment year, body mass index, other metabolic indices, smoking, alcohol drinking, and medication use, the hazard ratio (HR) for diabetes remission was 3.28 (95% CI: 2.75, 3.92; p < 0.001) for people with ≥10% weight loss within 1 year of diagnosis, 2.29 (95% CI: 2.03, 2.59; p < 0.001) for those with 5% to 9.9% weight loss, and 1.34 (95% CI: 1.22, 1.47; p < 0.001) for those with 0% to 4.9% weight loss compared to people with weight gain. During a median follow-up of 3.1 years, 67.2% (1,531) of people who had achieved diabetes remission returned to hyperglycaemia, with an incidence rate of 184.8 (95% CI: 175.5, 194.0) per 1,000 person-years. The adjusted HR for returning to hyperglycaemia was 0.52 (95% CI: 0.41, 0.65; p < 0.001) for people with ≥10% weight loss, 0.78 (95% CI: 0.68, 0.92; p = 0.002) for those with 5% to 9.9% weight loss, and 0.90 (95% CI: 0.80, 1.01; p = 0.073) for those with 0% to 4.9% weight loss compared to people with weight gain. Diabetes remission was associated with a 31% (HR: 0.69, 95% CI: 0.52, 0.93; p = 0.014) decreased risk of all-cause mortality. The main limitation of the study is that the reliability of HbA1c used to define diabetes remission can be affected by other medical conditions. Furthermore, we did not have data on bariatric surgery. CONCLUSIONS In this study, greater weight loss within the first year of diabetes diagnosis was associated with an increased likelihood of achieving diabetes remission and a decreased risk of returning to hyperglycaemia among those who had achieved diabetes remission. However, both the incidence of diabetes remission and the probability of its long-term sustainability were low with conventional management in real-world settings, in an era when the importance of weight loss was not fully appreciated. Our study provides evidence for policymakers to design and implement early weight management interventions and diabetes remission initiatives.
Collapse
Affiliation(s)
- Hongjiang Wu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Aimin Yang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
| | - Eric S. H. Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Xinge Zhang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Ronald C. W. Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Alice P. S. Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Wing-Yee So
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Hospital Authority, Hong Kong Special Administrative Region, China
| | - Juliana C. N. Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Andrea O. Y. Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
41
|
Park J, Ntelis S, Yunasan E, Downton KD, Yip TCF, Munir KM, Haq N. Glucagon-Like Peptide 1 Analogues as Adjunctive Therapy for Patients With Type 1 Diabetes: An Updated Systematic Review and Meta-analysis. J Clin Endocrinol Metab 2023; 109:279-292. [PMID: 37561012 DOI: 10.1210/clinem/dgad471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/24/2023] [Accepted: 08/09/2023] [Indexed: 08/11/2023]
Abstract
CONTEXT Concomitant obesity is common among patients with type 1 diabetes mellitus (T1DM), yet adjunctive therapy options are scarce. OBJECTIVE We assess the efficacy and adverse outcomes of glucagon-like peptide 1 (GLP-1) analogues when used as adjunctive therapy for T1DM. METHOD PubMed, EMBASE, Cochrane Central, and Scopus databases were searched for randomized controlled trials up to December 2022. Efficacy outcomes were A1c level, body weight, and total daily insulin (TDI) after ≥12 weeks of GLP-1 therapy. We also assessed 12 different adverse outcomes. Subgroup analysis was done for newly diagnosed or C-peptide positive (C-pos) patients. We report the certainty of evidence based on the GRADE assessment tool. RESULTS A total of 24 studies using 4 different GLP-1 analogues with a total of 3377 patients were included. Liraglutide had the most substantial evidence with effect sizes on A1c (-0.09%/mg), weight (-2.2 kg/mg), and TDI (-4.32 IU/mg). Liraglutide dose was the greatest predictor of greater average weight loss and TDI decrease but was associated with higher odds of nausea (OR 6.5; 95% CI, 5.0-8.4) and ketosis (OR 1.8; 95% CI, 1.1-2.8). Odds of severe (OR 0.67; 95% CI, 0.43-1.04) or symptomatic hypoglycemia (OR 0.89; 95% CI, 0.53-1.51) were not significantly elevated. Among C-pos patients, greater A1c decrease (-0.51% vs -0.28%) but similar weight loss and TDI were seen. Effect sizes for exenatide were similar, but studies had higher risk of bias and safety data were sparse. CONCLUSION Our meta-analysis supports therapeutic benefits of liraglutide for patients with T1DM mainly for weight loss and insulin dose reduction. Newly diagnosed or C-pos patients do not appear to experience greater weight loss benefits.
Collapse
Affiliation(s)
- Jeayoung Park
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Spyridon Ntelis
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Elvina Yunasan
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Katherine D Downton
- Health Sciences and Human Services Library, University of Maryland, Baltimore, MD 21201, USA
| | - Terry Cheuk-Fung Yip
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Central Ave, Hong Kong
| | - Kashif M Munir
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nowreen Haq
- Lead Endocrinologist, Diabetes and Obesity Management Clinic, Chase Brexton Healthcare, Baltimore, MD 21201, USA
| |
Collapse
|
42
|
Bariatric Surgery for Adults With Class I Obesity and Difficult-to-Manage Type 2 Diabetes: A Health Technology Assessment. ONTARIO HEALTH TECHNOLOGY ASSESSMENT SERIES 2023; 23:1-151. [PMID: 38130940 PMCID: PMC10732121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Background Many individuals with type 2 diabetes are classified as either overweight or obese. A patient may be described as having difficult-to-manage type 2 diabetes if their HbA1c levels remain above recommended target levels, despite efforts to treat it with lifestyle changes and pharmacotherapy. Bariatric surgery refers to procedures that modify the gastrointestinal tract. In patients with class II or III obesity, bariatric surgery has resulted in substantial weight loss, improved quality of life, reduced mortality risk, and resolution of type 2 diabetes. There is some evidence suggesting these outcomes may also be possible for patients with class I obesity as well. We conducted a health technology assessment of bariatric surgery for adults with class I obesity and difficult-to-manage type 2 diabetes, which included an evaluation of effectiveness, safety, cost-effectiveness, the budget impact of publicly funding bariatric surgery, and patient preferences and values. Methods We performed a systematic clinical literature review. We assessed the risk of bias of each included study, using the Cochrane Risk of Bias tool for randomized controlled trials, the Risk of Bias in Non-randomized Studies - of Interventions (ROBINS-I) tool for cohort studies, and the Risk of Bias in Systematic Reviews (ROBIS) tool for systematic reviews; we assessed the quality of the body of evidence according to the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) Working Group criteria. We performed a systematic economic literature review and conducted a cost-utility analysis of bariatric surgery in comparison with nonsurgical usual care over a lifetime horizon from a public payer perspective. We also analyzed the budget impact of publicly funding bariatric surgery for adults with class I obesity and difficult-to-manage type 2 diabetes in Ontario. To contextualize the potential value of bariatric surgery, we spoke with people with obesity and type 2 diabetes who had undergone or were considering this procedure. Results We included 14 studies in the clinical evidence review. There were large increases in diabetes remission rates (GRADE: Low to Very low) and large reductions in body mass index (GRADE: Low to Very low) with bariatric surgery than with medical management. Bariatric surgery may also reduce the use of medications for type 2 diabetes (GRADE: Low) and may improve quality of life for people with class I obesity and difficult-to-manage type 2 diabetes compared with medical management. (GRADE: Low)Our economic evidence review included 5 cost-effectiveness studies; none were conducted in a Canadian setting, and 4 were considered partially applicable to our research question. Most studies found bariatric surgery to be cost-effective compared to standard care for patients with class I obesity and type 2 diabetes; however, the applicability of these results to the Ontario context is uncertain due to potential differences in clinical practice, resource utilization, and unit costs.Our primary economic evaluation found that over a lifetime horizon, bariatric surgery was more costly (incremental cost: $8,151 per person) but also more effective than current usual care (led to a 0.339 quality-adjusted life-year [QALY] gain per person). The cost increase was driven by costs associated with surgery (before, after, and during surgery), and the QALY gain was due to life-years gained. Results were sensitive to the bariatric surgery cost and assumptions regarding its long-term benefits with respect to weight loss and diabetes remission.Publicly funding 50 bariatric surgeries in year 1, and gradually increasing to 250 surgeries in year 5, for people with class I obesity and difficult-to-manage type 2 diabetes would lead to budget increases of $0.55 million in year 1 to $2.45 million in year 5, for a total of $7.63 million over 5 years.The people with obesity and type 2 diabetes with whom we spoke reported that bariatric surgery was generally seen as a positive treatment option, and those who had undergone the procedure reported positively on its value as a treatment to manage their weight and diabetes. Conclusions For adults with class I obesity and difficult-to-manage type 2 diabetes, bariatric surgery may be more clinically effective and cost-effective than medical management. Compared with medical management in people with class I obesity and difficult-to-manage type 2 diabetes, bariatric surgery may result in large increases in diabetes remission rates, large reductions in BMI, and reduced medication use for type 2 diabetes, improved quality of life. Over a lifetime horizon, bariatric surgery led to a cost increase and QALY gain. Bariatric surgery can result in postsurgical complications that are not faced by those receiving medical management. The cost-effectiveness of bariatric surgery depends on its long-term impacts on obesity-related and diabetes-related complications, which could be uncertain.Our budget impact analysis suggests that publicly funding bariatric surgery in Ontario for people with class I obesity and difficult-to-manage type 2 diabetes would lead to a budget increase of $7.63 million over 5 years.For people with obesity and type 2 diabetes, bariatric surgery was seen as a potential positive treatment option to manage their weight and diabetes.
Collapse
|
43
|
Salehi M, Peterson R, Tripathy D, Pezzica S, DeFronzo R, Gastaldelli A. Differential effect of gastric bypass versus sleeve gastrectomy on insulinotropic action of endogenous incretins. Obesity (Silver Spring) 2023; 31:2774-2785. [PMID: 37853989 PMCID: PMC10593483 DOI: 10.1002/oby.23872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 10/20/2023]
Abstract
OBJECTIVE Prandial hyperinsulinemia after Roux-en-Y gastric bypass surgery (GB), and to lesser degree after sleeve gastrectomy (SG), has been attributed to rapid glucose flux from the gut and increased insulinotropic gut hormones. However, β-cell sensitivity to exogenous incretin is reduced after GB. This study examines the effect of GB versus SG on prandial glycemia and β-cell response to increasing concentrations of endogenous incretins. METHODS Glucose kinetics, insulin secretion rate (ISR), and incretin responses to 50-g oral glucose ingestion were compared between ten nondiabetic participants with GB versus nine matched individuals with SG and seven nonoperated normal glucose tolerant control individuals (CN) with and without administration of 200 mg of sitagliptin. RESULTS Fasting glucose and hormonal levels were similar among three groups. Increasing plasma concentrations of endogenous incretins by two- to three-fold diminished prandial glycemia and increased β-cell secretion in all three groups (p < 0.05), but insulin secretion per insulin sensitivity (i.e., disposition index) was increased only in GB (p < 0.05 for interaction). However, plot of the slope of ISR (from premeal to peak values) versus plasma glucagon-like peptide-1 concentration was smaller after GB compared with SG and CN. CONCLUSIONS After GB, increasing incretin activity augments prandial β-cell response whereas the β-cell sensitivity to increasing plasma concentrations of endogenous incretin is diminished.
Collapse
Affiliation(s)
- Marzieh Salehi
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
- STVHCS, Audie Murphy Hospital, San Antonio, TX, United States
| | - Richard Peterson
- Department of Surgery, University of Texas at San Antonio, San Antonio, TX, United States
| | - Devjit Tripathy
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
| | - Samantha Pezzica
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| | - Ralph DeFronzo
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
| | - Amalia Gastaldelli
- Division of Diabetes, University of Texas at San Antonio, San Antonio, TX, United States
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| |
Collapse
|
44
|
Zhou X, Ke Z, Sun F, Li F, You Z, Zhang J, Gao Y, Zhu Z, Tong W. Effect of Bariatric Surgery on Metabolic Syndrome in Chinese Patients with Low Body Mass Index: a Propensity Score Matching Study. Obes Surg 2023; 33:3051-3061. [PMID: 37561268 DOI: 10.1007/s11695-023-06765-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 08/11/2023]
Abstract
BACKGROUND Bariatric surgery (BS) is known to improve the components of metabolic syndrome (MS) in patients with obesity. However, few studies have evaluated the effect of BS on patients with MS at low BMI levels. The study aims to assess the effect of BS on MS in patients with low BMI by comparing BS with medical therapy (MT). METHODS A total of 271 patients with MS who underwent BS and MT at a single institution were reviewed in this retrospective cohort study. A 1:1 propensity score matching was performed for the BS and MT groups with BMI<35 kg/m2. We analyzed the 5 years effect of BS versus MT on the remission of MS, its components, atherosclerotic cardiovascular disease (ASCVD) risk, and medication used. RESULTS Patients in the MT group showed a decrease in the prevalence of MS to 62% at the 1st year and 10% at the 5th year. In the BS group, MS prevalence in patients with BMI<35 kg/m2 decreased to 30% and 9% at the 1st and 5th year, whereas in patients with BMI≥35 kg/m2 was 26% and 7%, respectively. The 10-year ASCVD risk and the lifetime ASCVD were significantly decreased 5 years after BS in patients with BMI<35 kg/m2. The number and daily drug dose of hypoglycemic drugs, antihypertensive medications, and lipid-lowering drugs were reduced from baseline in both BS and MT groups. CONCLUSION Bariatric surgery significantly improves MS remission rates and long-term cardiovascular risk in Chinese patients with metabolic syndrome and a BMI <35 kg/m2.
Collapse
Affiliation(s)
- Xunmei Zhou
- Department of Endocrinology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, China
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Zhigang Ke
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China
| | - Fan Li
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zaichun You
- Department of General Medicine, Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Jian Zhang
- Department of General Medicine, Xinmiao Central Hospital, Fuling District, Chongqing, 408114, China
| | - Yu Gao
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Army Medical University, Chongqing Institute of Hypertension, Chongqing, 400042, China.
| | - Weidong Tong
- Department of General Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
45
|
Salehi M, Tripathy D, Peterson R, Honka H, Pezzica S, DeFronzo R, Gastaldelli A. Bariatric Surgery Alters the Postprandial Recovery From Hypoglycemia, Mediated by Cholinergic Signal. Diabetes 2023; 72:1374-1383. [PMID: 37467435 PMCID: PMC10545558 DOI: 10.2337/db23-0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/03/2023] [Indexed: 07/21/2023]
Abstract
Roux-en-Y gastric bypass (GB) and sleeve gastrectomy (SG) surgeries increase prandial insulin and glucagon secretion but reduce the endogenous glucose production (EGP) response to hypoglycemia in comparison with control subjects who had not undergone gastric surgery (CN), suggesting that parasympathetic nervous system (PNS) plays a role. Here, we investigated the effect of acute PNS blockade on the post-meal counterregulatory response to insulin-induced hypoglycemia in GB and SG compared with CN. Glucose kinetics and islet cell secretion were measured in nine subjects without diabetes with GB and seven with SG and five CN during hyperinsulinemic-hypoglycemic clamp (∼3.2 mmol/L) combined with meal ingestion on two separate days with and without intravenous atropine infusion. Glucose and hormonal levels were similar at baseline and during steady-state hypoglycemia before meal ingestion in three groups and unaffected by atropine. Atropine infusion diminished prandial systemic appearance of ingested glucose (RaO) by 30%, EGP by 40%, and glucagon response to hypoglycemia by 90% in CN. In GB or SG, blocking PNS had no effect on the RaO or meal-induced hyperglucagonemia but increased EGP in SG without any effect in GB (P < 0.05 interaction). These findings indicate that cholinergic signal contributes to the recovery from hypoglycemia by meal consumption in humans. However, bariatric surgery dissipates PNS-mediated physiologic responses to hypoglycemia in the fed state. ARTICLE HIGHLIGHTS Rerouted gut after Roux-en-Y gastric bypass (GB) and, to a lesser degree, after sleeve gastrectomy (SG) leads to larger glucose excursion and lower nadir glucose, predisposing individuals to hypoglycemia. Despite prandial hyperglucagonemia, endogenous glucose production response to hypoglycemia is reduced after GB or SG. Parasympathetic nervous system (PNS) activity plays a key role in regulation of glucose kinetics and islet cell function. We examined the effect of acute PNS blockade on counterregulatory glucose and islet cell response to meal ingestion during insulin-induced hypoglycemia among GB, SG, and control subjects who had not had gastric surgery. Our findings demonstrate that cholinergic signal is critical in the recovery from hypoglycemia by meal ingestion in humans who have not had gastric surgery, although prandial PNS-mediated physiologic responses to hypoglycemia are differentially changed by GB and SG.
Collapse
Affiliation(s)
- Marzieh Salehi
- Division of Diabetes, The University of Texas at San Antonio, San Antonio, TX
- Audie L. Murphy Memorial Veterans’ Hospital, South Texas Veterans Health Care System, San Antonio, TX
| | - Devjit Tripathy
- Division of Diabetes, The University of Texas at San Antonio, San Antonio, TX
| | - Richard Peterson
- Department of Surgery, The University of Texas at San Antonio, San Antonio, TX
| | - Henri Honka
- Division of Diabetes, The University of Texas at San Antonio, San Antonio, TX
| | - Samantha Pezzica
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| | - Ralph DeFronzo
- Division of Diabetes, The University of Texas at San Antonio, San Antonio, TX
| | - Amalia Gastaldelli
- Division of Diabetes, The University of Texas at San Antonio, San Antonio, TX
- Cardiometabolic Risk Unit, CNR Institute of Clinical Physiology, Pisa, Italy
| |
Collapse
|
46
|
Sayyed Kassem L, Rajpal A, Barreiro MV, Ismail‐Beigi F. Beta-cell function in type 2 diabetes (T2DM): Can it be preserved or enhanced? J Diabetes 2023; 15:817-837. [PMID: 37522521 PMCID: PMC10590683 DOI: 10.1111/1753-0407.13446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 05/26/2023] [Accepted: 06/11/2023] [Indexed: 08/01/2023] Open
Abstract
Type 2 diabetes (T2DM) is a complex metabolic disorder manifested by hyperglycemia, insulin resistance, and deteriorating beta-cell function. A way to prevent progression of the disease might be to enhance beta-cell function and insulin secretion. However, most previous studies examined beta-cell function while patients were using glycemia-lowering agents without an adequate period off medications (washout). In the present review we focus on studies with a washout period. We performed a literature search (2010 to June 2021) using beta-cell function and enhancement. The evidence shows that beta-cell function can be enhanced. Bariatric surgery and very low calorie diets show improvement in beta-cell function in many individuals. In addition, use of glucagon-like peptide-1 receptor agonists for prolonged periods (3 years or more) can also lead to improvement of beta-cell function. Further research is needed to understand the mechanisms leading to improved beta-cell function and identify agents that could enhance beta-cell function in patients with T2DM.
Collapse
Affiliation(s)
- Laure Sayyed Kassem
- Case Western Reserve UniversityClevelandOhioUSA
- Cleveland VA Medical CenterCase Western Reserve UniversityClevelandOhioUSA
| | - Aman Rajpal
- Case Western Reserve UniversityClevelandOhioUSA
- Cleveland VA Medical CenterCase Western Reserve UniversityClevelandOhioUSA
| | | | - Faramarz Ismail‐Beigi
- Case Western Reserve UniversityClevelandOhioUSA
- Cleveland VA Medical CenterCase Western Reserve UniversityClevelandOhioUSA
- University Hospitals of ClevelandClevelandOhioUSA
| |
Collapse
|
47
|
Ye J, Hu Y, Wang C, Lian H, Dong Z. Cellular mechanism of diabetes remission by bariatric surgery. Trends Endocrinol Metab 2023; 34:590-600. [PMID: 37574405 DOI: 10.1016/j.tem.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/13/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023]
Abstract
Bariatric surgery is a powerful therapy for type 2 diabetes in patients with obesity. The mechanism of insulin sensitization by surgery has been extensively investigated in weight loss-dependent and weight loss-independent conditions. However, a consensus remains to be established regarding the underlying mechanisms. Energy deficit induced by calorie restriction (CR), that occurs both before and after surgery, represents a unique physiological basis for insulin sensitization regardless of weight loss. In support, we integrate evidence in the literature to provide an energy-based view of insulin sensitization as follows: surgery improves insulin sensitivity through the energy deficit induced by CR, leading to correction of mitochondrial overload in multiple cell types; this then triggers functional reprogramming of relevant tissues leading to diabetes remission.
Collapse
Affiliation(s)
- Jianping Ye
- Metabolic Disease Research Center, Zhengzhou University Affiliated Zhengzhou Central Hospital, Zhengzhou 450007, China; Center for Advanced Medicine, College of Medicine, Zhengzhou University, Zhengzhou 450052, China; Research Center for Basic Medicine, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Yangxi Hu
- Department of Metabolic Surgery, Zhengzhou University Affiliated Zhengzhou Central Hospital, Zhengzhou 450007, China
| | - Chengming Wang
- Metabolic Disease Research Center, Zhengzhou University Affiliated Zhengzhou Central Hospital, Zhengzhou 450007, China
| | - Hongkai Lian
- Trauma Research Center, Zhengzhou University Affiliated Zhengzhou Central Hospital, Zhengzhou 450007, China
| | - Zigang Dong
- Center for Advanced Medicine, College of Medicine, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
48
|
Chang YC, Hsu CN, Chong K, Yang PJ, Ser KH, Lee PC, Chen SC, Hsuan CF, Lee YC, Hsu CC, Lee HL, Liao KCW, Hsieh ML, Chuang GT, Yang WS, Chu SL, Li WY, Chuang LM, Lee WJ. Roux-en-Y and One-Anastomosis Gastric Bypass Surgery Are Superior to Sleeve Gastrectomy in Lowering Glucose and Cholesterol Levels Independent of Weight Loss: a Propensity-Score Weighting Analysis. Obes Surg 2023; 33:3035-3050. [PMID: 37612578 DOI: 10.1007/s11695-023-06656-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND The superior effects of gastric bypass surgery in preventing cardiovascular diseases compared with sleeve gastrectomy are well-established. However, whether these effects are independent of weight loss is not known. METHODS In this retrospective cohort study, we compared the change in cardiometabolic risks of 1073 diabetic patients undergoing Roux-en-Y gastric bypass (RYGB) (n = 265), one-anastomosis gastric bypass (OAGB) (n = 619), and sleeve gastrectomy (SG) (n = 189) with equivalent weight loss from the Min-Shen General Hospital. Propensity score-weighting, multivariate regression, and matching were performed to adjust for baseline differences. RESULTS After 12 months, OAGB and, to a lesser extent, RYGB exhibited superior effects on glycemic control compared with SG in patients with equivalent weight loss. The effect was significant in patients with mild-to-modest BMI reduction but diminished in patients with severe BMI reduction. RYGB and OAGB had significantly greater effects in lowering total and low-density lipoprotein cholesterol than SG, regardless of weight loss. The results of matching patients with equivalent weight loss yielded similar results. The longer length of bypassed biliopancreatic (BP) limbs was correlated with a greater decrease in glycemic levels, insulin resistance index, lipids, C-reactive protein (CRP) levels, and creatinine levels in patients receiving RYBG. It was correlated with greater decreases in BMI, fasting insulin, insulin resistance index, and C-reactive protein levels in patients receiving OAGB. CONCLUSION Diabetic patients receiving OAGB and RYGB had lower glucose and cholesterol levels compared with SG independent of weight loss. Our results suggest diabetic patients with cardiovascular risk factors such as hypercholesterolemia to receive bypass surgery.
Collapse
Affiliation(s)
- Yi-Cheng Chang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Center for Obesity, Lifestyle, and Metabolic Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
- Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Chih-Neng Hsu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, 640, Taiwan
| | - Keong Chong
- Department of Medicine, Min-Sheng General Hospital, Taoyuan, 330, Taiwan
| | - Po-Jen Yang
- Center for Obesity, Lifestyle, and Metabolic Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
- Department of Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Kong-Han Ser
- Department of Surgery, Ten-Chen General Hospital, Taoyuan, 326, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Shu-Chun Chen
- Department of Nursing, Ming-Sheng General Hospital, Taoyuan, 330, Taiwan
| | - Chin-Feng Hsuan
- Department of Internal Medicine, Division of Cardiology, E-Da Hospital, Kaohsiung, 824, Taiwan
- Department of Internal Medicine, Division of Cardiology, E-Da Dachang Hospital, Kaohsiung, 824, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 840, Taiwan
| | - Yi-Chih Lee
- Department of International Business, Chien Hsin University of Science and Technology, Taoyuan, 320, Taiwan
| | - Chih-Cheng Hsu
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, 350, Taiwan
| | - Hsiao-Lin Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Karen Chia-Wen Liao
- Biological Sciences Division, University of Chicago, Chicago, IL, 60637, USA
| | - Meng-Lun Hsieh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Gwo-Tsann Chuang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100, Taiwan
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Wei-Shun Yang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, 302, Taiwan
| | - Shao-Lun Chu
- Department of Medicine, College of Medicine, National Taiwan University, Taipei, 100, Taiwan
| | - Wen-Yi Li
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, 640, Taiwan
| | - Lee-Ming Chuang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan.
- Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, 100, Taiwan.
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, 100, Taiwan.
| | - Wei-Jei Lee
- Department of Medicine, Min-Sheng General Hospital, Taoyuan, 330, Taiwan.
- Department of Surgery, Min-Sheng General Hospital, Taoyuan, 330, Taiwan.
| |
Collapse
|
49
|
Orioli L, Canouil M, Sawadogo K, Ning L, Deldicque L, Lause P, de Barsy M, Froguel P, Loumaye A, Deswysen Y, Navez B, Bonnefond A, Thissen JP. Identification of myokines susceptible to improve glucose homeostasis after bariatric surgery. Eur J Endocrinol 2023; 189:409-421. [PMID: 37638789 DOI: 10.1093/ejendo/lvad122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023]
Abstract
IMPORTANCE AND OBJECTIVE The identification of myokines susceptible to improve glucose homeostasis following bariatric surgery could lead to new therapeutic approaches for type 2 diabetes. METHODS Changes in the homeostasis model assessment (HOMA) test were assessed in patients before and 3 months after bariatric surgery. Changes in myokines expression and circulating levels were assessed using real-time quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). Myokines known to regulate glucose homeostasis were identified using literature (targeted study) and putative myokines using RNA-sequencing (untargeted study). A linear regression analysis adjusted for age and sex was used to search for associations between changes in the HOMA test and changes in myokines. RESULTS In the targeted study, brain-derived neurotrophic factor (BDNF) expression was upregulated (+30%, P = .006) while BDNF circulating levels were decreased (-12%, P = .001). Upregulated BDNF expression was associated with decreased HOMA of insulin resistance (HOMA-IR) (adjusted estimate [95% confidence interval {CI}]: -0.51 [-0.88 to -0.13], P = .010). Decreased BDNF serum levels were associated with decreased HOMA of beta-cell function (HOMA-B) (adjusted estimate [95% CI] = 0.002 [0.00002-0.0031], P = .046). In the untargeted study, upregulated putative myokines included XYLT1 (+64%, P < .001), LGR5 (+57, P< .001), and SPINK5 (+46%, P < .001). Upregulated LGR5 was associated with decreased HOMA-IR (adjusted estimate [95% CI] = -0.50 [-0.86 to -0.13], P = .009). Upregulated XYLT1 and SPINK5 were associated with increased HOMA of insulin sensitivity (HOMA-S) (respectively, adjusted estimate [95% CI] = 109.1 [28.5-189.8], P = .009 and 16.5 [0.87-32.19], P = .039). CONCLUSIONS Improved glucose homeostasis following bariatric surgery is associated with changes in myokines expression and circulating levels. In particular, upregulation of BDNF, XYLT1, SPINK5, and LGR5 is associated with improved insulin sensitivity. These results suggest that these myokines could contribute to improved glucose homeostasis following bariatric surgery. STUDY REGISTRATION NCT03341793 on ClinicalTrials.gov (https://clinicaltrials.gov/).
Collapse
Affiliation(s)
- Laura Orioli
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Mickaël Canouil
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Kiswendsida Sawadogo
- Statistical Support Unit, King Albert II Cancer and Hematology Institute, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Lijiao Ning
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Louise Deldicque
- Institute of NeuroScience, Université Catholique de Louvain, 1348 Louvain-La-Neuve, Belgium
| | - Pascale Lause
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Marie de Barsy
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Philippe Froguel
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, London SW7 2BX, United Kingdom
| | - Audrey Loumaye
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Yannick Deswysen
- Department of Oeso-gastro-duodenal and Bariatric Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Benoit Navez
- Department of Oeso-gastro-duodenal and Bariatric Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Amélie Bonnefond
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Jean-Paul Thissen
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
50
|
Haran A, Bergel M, Kleiman D, Hefetz L, Israeli H, Weksler-Zangen S, Agranovich B, Abramovich I, Ben-Haroush Schyr R, Gottlieb E, Ben-Zvi D. Differential effects of bariatric surgery and caloric restriction on hepatic one-carbon and fatty acid metabolism. iScience 2023; 26:107046. [PMID: 37389181 PMCID: PMC10300224 DOI: 10.1016/j.isci.2023.107046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/24/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023] Open
Abstract
Weight loss interventions, including dietary changes, pharmacotherapy, or bariatric surgery, prevent many of the adverse consequences of obesity, and may also confer intervention-specific benefits beyond those seen with decreased weight alone. We compared the molecular effects of different interventions on liver metabolism to understand the mechanisms underlying these benefits. Male rats on a high-fat, high-sucrose diet underwent sleeve gastrectomy (SG) or intermittent fasting with caloric restriction (IF-CR), achieving equivalent weight loss. The interventions were compared to ad-libitum (AL)-fed controls. Analysis of liver and blood metabolome and transcriptome revealed distinct and sometimes contrasting metabolic effects between the two interventions. SG primarily influenced one-carbon metabolic pathways, whereas IF-CR increased de novo lipogenesis and glycogen storage. These findings suggest that the unique metabolic pathways affected by SG and IF-CR contribute to their distinct clinical benefits, with bariatric surgery potentially influencing long-lasting changes through its effect on one-carbon metabolism.
Collapse
Affiliation(s)
- Arnon Haran
- Department of Hematology, Haddasah Medical Center, Jerusalem, Israel
| | - Michael Bergel
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Doron Kleiman
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Liron Hefetz
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Hadar Israeli
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | | | - Bella Agranovich
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ifat Abramovich
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Rachel Ben-Haroush Schyr
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Eyal Gottlieb
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Danny Ben-Zvi
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, The Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|